Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Stem Cell Rev Rep ; 10(5): 712-24, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24906426

ABSTRACT

Amniotic fluid (AF) is a source of multipotent mesenchymal stem cells (MSCs), very promising cells for tissue engineering in clinical application. The aim of this work was to isolate and characterize cells isolated from bovine AF as alternative sources of primitive multipotent stem cells in a species that could be a large-animal model for biomedical and biotechnology researches. Samples were recovered, at slaughterhouse, from 39 pregnant cows at different trimesters of pregnancy and cells were cultured in vitro. At passages (P) 3 and 7 differentiation towards chondrogenic, osteogenic and adipogenic lineages was induced. Flow cytometry analysis for CD90, CD105, CD73, CD44, CD34, CD45 and CD14 was performed, immunocytochemistry (ICC) for Oct4, SSEA4, α-SMA, Vimentin, N- and E- Cadherin and CK and qPCR analysis for OCT4, NANOG and SOX2 were carried out. The cell yield was significantly higher in the first trimester compared to the second and the third one (P < 0.05). Cells were isolated from 25/39 samples and cell population appeared heterogeneous. Two main cell types were identified in samples from all trimesters: round- (RS) and spindle-shaped (SS) cells. 17/25 samples showed both populations (mixed, MX). Both cell types showed MSC-markers and differentiation capability with some variability related to the passages. The SS-population also expressed low levels of stemness markers such as NANOG and SSEA4 but not OCT4. Bovine AF shows a heterogeneous cell population containing also MSCs, multipotent cells that represent an intermediate stage between embryonic stem cells and adult ones.


Subject(s)
Amniotic Fluid/cytology , Mesenchymal Stem Cells/cytology , Pregnancy, Animal/physiology , Animals , Cattle , Cell Differentiation , Cell Separation , Cells, Cultured , Female , Flow Cytometry , Multipotent Stem Cells/cytology , Pregnancy
2.
Leukemia ; 28(8): 1687-97, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24480986

ABSTRACT

Peripheral T-cell lymphomas not otherwise specified (PTCL/NOS) are very aggressive tumors characterized by consistent aberrant expression of platelet-derived growth factor receptor alpha (PDGFRA). In this study, we aimed to identify the determinants of PDGFRA activity in PTCL/NOS and to elucidate the biological consequences of its activation. We observed overexpression of the PDGFRA gene by gene expression profiling in most of the tested PTCLs and confirmed the expression of PDGFRA and phospho-PDGFRA using immunohistochemistry. The integrity of the PDFGRA locus was demonstrated using several different approaches, including massive parallel sequencing and Sanger sequencing. PDGF-AA was found to be expressed and secreted by PTCL/NOS cells and to be necessary and sufficient for PDGFRA phosphorylation ex vivo by sustaining an autocrine stimulation. We documented consistently high PDGF-A expression in primary biopsies and patients' plasma and tracked PDGFRA signaling in primary tumors, achieving evidence of its activation. Indeed, we found that STAT1 and STAT5 are implicated in PDGFRA signaling transduction. Finally, we demonstrated that PDGFRA activation supported tumor cell proliferation and provided the first evidence of the anti-lymphoma activity of PDGRA inhibition in a PTCL/NOS patient. Altogether, our data demonstrated that PDGFRA activity fosters PTCL/NOS proliferation via an autocrine loop.


Subject(s)
Autocrine Communication , Lymphoma, T-Cell, Peripheral/pathology , Platelet-Derived Growth Factor/physiology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Proto-Oncogene Proteins c-akt/physiology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , STAT1 Transcription Factor , STAT5 Transcription Factor/physiology
3.
Leukemia ; 28(6): 1196-206, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24310736

ABSTRACT

Constitutively active phosphoinositide 3-kinase (PI3K) signaling is a common feature of T-cell acute lymphoblastic leukemia (T-ALL), where it upregulates cell proliferation, survival and drug resistance. These observations lend compelling weight to the application of PI3K inhibitors in the therapy of T-ALL. Here, we have analyzed the therapeutic potential of the pan-PI3K inhibitor NVP-BKM120 (BKM120), an orally bioavailable 2,6-dimorpholino pyrimidine derivative, which has entered clinical trials for solid tumors, on both T-ALL cell lines and patient samples. BKM120 treatment resulted in G2/M phase cell cycle arrest and apoptosis, being cytotoxic to a panel of T-ALL cell lines and patient T lymphoblasts, and promoting a dose- and time-dependent dephosphorylation of Akt and S6RP. BKM120 maintained its pro-apoptotic activity against Jurkat cells even when cocultured with MS-5 stromal cells, which mimic the bone marrow microenvironment. Remarkably, BKM120 synergized with chemotherapeutic agents currently used for treating T-ALL patients. Moreover, in vivo administration of BKM120 to a subcutaneous xenotransplant model of human T-ALL significantly delayed tumor growth, thus prolonging survival time. Taken together, our findings indicate that BKM120, either alone or in combination with chemotherapeutic drugs, may be an efficient treatment for T-ALLs that have aberrant upregulation of the PI3K signaling pathway.


Subject(s)
Aminopyridines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Blotting, Western , Flow Cytometry , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Leukemia ; 28(4): 739-48, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23892718

ABSTRACT

B-precursor acute lymphoblastic leukemia (B-pre ALL) is a malignant disorder characterized by the abnormal proliferation of B-cell progenitors. The prognosis of B-pre ALL has improved in pediatric patients, but the outcome is much less successful in adults. Constitutive activation of the phosphatidylinositol 3-kinase (PI3K), Akt and the mammalian target of rapamycin (mTOR) (PI3K/Akt/mTOR) network is a feature of B-pre ALL, where it strongly influences cell growth and survival. RAD001, a selective mTORC1 inhibitor, has been shown to be cytotoxic against many types of cancer including hematological malignancies. To investigate whether mTORC1 could represent a target in the therapy of B-pre ALL, we treated cell lines and adult patient primary cells with RAD001. We documented that RAD001 decreased cell viability, induced cell cycle arrest in G0/G1 phase and caused apoptosis in B-pre ALL cell lines. Autophagy was also induced, which was important for the RAD001 cytotoxic effect, as downregulation of Beclin-1 reduced drug cytotoxicity. RAD001 strongly synergized with the novel allosteric Akt inhibitor MK-2206 in both cell lines and patient samples. Similar results were obtained with the combination CCI-779 plus GSK 690693. These findings point out that mTORC1 inhibitors, either as a single agent or in combination with Akt inhibitors, could represent a potential therapeutic innovative strategy in B-pre ALL.


Subject(s)
Phosphoinositide-3 Kinase Inhibitors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Everolimus , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/antagonists & inhibitors , Oxadiazoles/pharmacology , Sirolimus/analogs & derivatives , Sirolimus/pharmacology
5.
Vox Sang ; 105(2): 116-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23600766

ABSTRACT

BACKGROUND AND OBJECTIVES: The haemoglobin level of prospective blood donors is usually performed on blood obtained by from the finger pulp by fingerstick with a lancet and filling a capillary tube with a sample. New noninvasive methods are now available for rapid, noninvasive predonation haemoglobin screening. MATERIALS AND METHODS: Prospective blood donors at our blood centre were tested, in two different trials, as follows: by the NBM 200 (OrSense) test (n = 445 donors) and by the Pronto-7 (Masimo) test (n = 463 donors). The haemoglobin values of each trial and the haemoglobin of finger pulp blood obtained by fingerstick with a lancet (HemoCue) were compared with the haemoglobin values obtained from a venous sample on a Cell Counter (Beckman Coulter). RESULTS: Comparison of Beckman Coulter Cell Counter and OrSense and results showed a bias of 0.29 g/dl, the standard deviation of the differences (SDD) of 0.98 and 95% limits of agreement from -1.64 to 2.21, using Bland and Altman statistical methodology. Comparison of Masimo and Beckman Coulter Cell Counter results showed a bias of -0.53 g/dl, SDD of 1.04 and 95% limits of agreement from -2.57 to 1.51. Cumulative analysis of all 908 donors, as tested by the usual fingerstick test showed a bias of 0.83 g/dl, SDD of 0.70 and 95% limits of agreement from -0.54 to 2.20 compared with the Coulter Cell Counter. Compared with the Coulter Counter, the specificity of the methods was 99.5% for fingerstick, 97% for OrSense and 83% for Massimo, and the sensitivity was 99, 98 and 93%, respectively. CONCLUSIONS: Analysis of finger pulp blood by either direct sampling by fingerstick and Hemocue, or by noninvasive haemoglobin tests does not replicate the results of cell counter analysis of venous samples. Compared with fingerstick, noninvasive haemoglobin tests eliminate pain and reduce stress, but have a lower level of specificity and sensitivity.


Subject(s)
Blood Chemical Analysis/instrumentation , Blood Chemical Analysis/methods , Blood Donors , Hemoglobins/metabolism , Female , Hemoglobins/analysis , Humans , Male , Prospective Studies
6.
Leukemia ; 26(11): 2336-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22614243

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive neoplastic disorder arising from T-cell progenitors. T-ALL accounts for 15% of newly diagnosed ALL cases in children and 25% in adults. Although the prognosis of T-ALL has improved, due to the use of polychemotherapy schemes, the outcome of relapsed/chemoresistant T-ALL cases is still poor. A signaling pathway that is frequently upregulated in T-ALL, is the phosphatidylinositol 3-kinase/Akt/mTOR network. To explore whether Akt could represent a target for therapeutic intervention in T-ALL, we evaluated the effects of the novel allosteric Akt inhibitor, MK-2206, on a panel of human T-ALL cell lines and primary cells from T-ALL patients. MK-2206 decreased T-ALL cell line viability by blocking leukemic cells in the G(0)/G(1) phase of the cell cycle and inducing apoptosis. MK-2206 also induced autophagy, as demonstrated by an increase in the 14-kDa form of LC3A/B. Western blotting analysis documented a concentration-dependent dephosphorylation of Akt and its downstream targets, GSK-3α/ß and FOXO3A, in response to MK-2206. MK-2206 was cytotoxic to primary T-ALL cells and induced apoptosis in a T-ALL patient cell subset (CD34(+)/CD4(-)/CD7(-)), which is enriched in leukemia-initiating cells. Taken together, our findings indicate that Akt inhibition may represent a potential therapeutic strategy in T-ALL.


Subject(s)
Heterocyclic Compounds, 3-Ring/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Blotting, Western , Cell Cycle/drug effects , Doxorubicin/pharmacology , Drug Synergism , Humans , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Signal Transduction
7.
Bone Marrow Transplant ; 47(8): 1105-11, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22101198

ABSTRACT

Several studies have shown that chronic GVHD (cGVHD) is more frequent in patients receiving transplants from PBSC than in those receiving BM. In the setting of PBSC-unrelated transplants, the addition of anti-T-cell globulin (ATG) has shown a significant decrease in incidence/severity of cGVHD, without an increase in relapses or infections. However, no prospective data are yet available in the sibling setting. We retrospectively analyzed the effects of intensification of standard GVHD prophylaxis (CsA+MTX) by the addition of low-dose ATG in 245 patients receiving a transplant from HLA-identical sibling. From 1996 to 2001, patients received PBSC as the preferred source (group 2), and then ATG was added before transplant (group 3) because of a high cGVHD rate. Patients receiving BM in the same time period were analyzed as a control group (group 1). The incidence of grade III-IV acute GVHD and cGVHD was not significantly different in the three groups, but extensive cGVHD was highest in group 2 (38%) compared with group 3 (21%) or group 1 (28%; P=0.03). OS, TRM and time to relapse/progression were similar in the three groups. Our analysis shows that adding ATG to PBSC sibling allogeneic transplants can lower cGVHD, without an increase of relapse. Further prospective studies are needed to confirm these findings.


Subject(s)
Antilymphocyte Serum/administration & dosage , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Immunosuppressive Agents/administration & dosage , Peripheral Blood Stem Cell Transplantation , Siblings , Acute Disease , Adolescent , Adult , Aged , Chronic Disease , Female , Graft vs Host Disease/etiology , Histocompatibility Testing , Humans , Male , Middle Aged , Retrospective Studies , Transplantation, Homologous
8.
Leukemia ; 26(1): 91-100, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21968881

ABSTRACT

The mammalian target of rapamycin (mTOR) serine/threonine kinase is the catalytic subunit of two multi-protein complexes, referred to as mTORC1 and mTORC2. Signaling downstream of mTORC1 has a critical role in leukemic cell biology by controlling mRNA translation of genes involved in both cell survival and proliferation. mTORC1 activity can be downmodulated by upregulating the liver kinase B1/AMP-activated protein kinase (LKB1/AMPK) pathway. Here, we have explored the therapeutic potential of the anti-diabetic drug, metformin (an LKB1/AMPK activator), against both T-cell acute lymphoblastic leukemia (T-ALL) cell lines and primary samples from T-ALL patients displaying mTORC1 activation. Metformin affected T-ALL cell viability by inducing autophagy and apoptosis. However, it was much less toxic against proliferating CD4(+) T-lymphocytes from healthy donors. Western blot analysis demonstrated dephosphorylation of mTORC1 downstream targets. Unlike rapamycin, we found a marked inhibition of mRNA translation in T-ALL cells treated with metformin. Remarkably, metformin targeted the side population of T-ALL cell lines as well as a putative leukemia-initiating cell subpopulation (CD34(+)/CD7(-)/CD4(-)) in patient samples. In conclusion, metformin displayed a remarkable anti-leukemic activity, which emphasizes future development of LKB1/AMPK activators as clinical candidates for therapy in T-ALL.


Subject(s)
Adenylate Kinase/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proteins/metabolism , Signal Transduction , Apoptosis , Base Sequence , Cell Line, Tumor , DNA Primers , Flow Cytometry , Humans , Mechanistic Target of Rapamycin Complex 1 , Metformin/pharmacology , Multiprotein Complexes , Phosphorylation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Biosynthesis/drug effects , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , TOR Serine-Threonine Kinases
9.
Leukemia ; 25(5): 781-91, 2011 May.
Article in English | MEDLINE | ID: mdl-21331075

ABSTRACT

The mammalian Target Of Rapamycin (mTOR) serine/threonine kinase belongs to two multi-protein complexes, referred to as mTORC1 and mTORC2. mTOR-generated signals have critical roles in leukemic cell biology by controlling mRNA translation of genes that promote proliferation and survival. However, allosteric inhibition of mTORC1 by rapamycin has only modest effects in T-cell acute lymphoblastic leukemia (T-ALL). Recently, ATP-competitive inhibitors specific for the mTOR kinase active site have been developed. In this study, we have explored the therapeutic potential of active-site mTOR inhibitors against both T-ALL cell lines and primary samples from T-ALL patients displaying activation of mTORC1 and mTORC2. The inhibitors affected T-ALL cell viability by inducing cell-cycle arrest in G(0)/G(1) phase, apoptosis and autophagy. Western blot analysis demonstrated a Ser 473 Akt dephosphorylation (indicative of mTORC2 inhibition) and a dephosphorylation of mTORC1 downstream targets. Unlike rapamycin, we found a marked inhibition of mRNA translation in T-ALL cell lines treated with active-site mTOR inhibitors. The inhibitors strongly synergized with both vincristine and the Bcl-2 inhibitor, ABT-263. Remarkably, the drugs targeted a putative leukemia-initiating cell sub-population (CD34(+)/CD7(-)/CD4(-)) in patient samples. In conclusion, the inhibitors displayed remarkable anti-leukemic activity, which emphasizes their future development as clinical candidates for therapy in T-ALL.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Animals , Blotting, Western , Catalytic Domain , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Humans , Immunosuppressive Agents/pharmacology , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , Phosphorylation/drug effects , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proteins/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transcription Factors/metabolism
10.
Eur Cell Mater ; 20: 13-23, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20597062

ABSTRACT

Platelet-rich plasma (PRP) is used clinically in liquid or gel form to promote tissue repair. Because of the poor mechanical properties, conventional PRP is often difficult to handle when used in clinical settings and requires secure implantation in a specific site, otherwise when released growth factors could be washed out during an operation. In this study, we analyzed the end product of a recently developed commercially available system (FIBRINET), which is a dense pliable, platelet-rich fibrin matrix (PRFM). We characterized the mechanical properties of PRFM and tested whether PRFM releases growth factors and whether released factors induce the proliferation of mesenchymal stem cells (MSC). Mechanical properties as well as platelet distribution were evaluated in PRFM. PRFM demonstrated robust mechanical properties, with a tear elastic modulus of 937.3 +/- 314.6 kPa, stress at a break of 1476.0 +/- 526.3 kPa, and an elongation at break of 146.3 +/- 33.8 %. PRFM maintained its mechanical properties throughout the testing process. Microscopic observations showed that the platelets were localized on one side of the matrix. Elevated levels of PDGF-AA, PDGF-AB, EGF, VEGF, bFGF and TGF-beta1 were measured in the day 1-conditioned media (CM) of PRFM and growth factor levels decreased thereafter. BMP2 and BMP7 were not detectable. MSC culture media supplemented with 20% PRFM-CM stimulated MSC cell proliferation; at 24 and 48 hours the induction of the proliferation was significantly greater than the induction obtained with media supplemented with 20% foetal bovine serum. The present study shows that the production of a dense, physically robust PRFM made through high-speed centrifugation of intact platelets and fibrin in the absence of exogenous thrombin yields a potential tool for accelerating tissue repair.


Subject(s)
Fibrin/metabolism , Platelet-Rich Plasma/metabolism , Cell Proliferation , Culture Media, Conditioned , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Platelet-Derived Growth Factor/metabolism , Platelet-Rich Plasma/cytology
11.
Eur J Vasc Endovasc Surg ; 40(3): 365-74, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20570185

ABSTRACT

OBJECTIVES AND DESIGN: To establish whether in diabetic patients with peripheral artery obstructive disease (PAOD) vasa vasorum (vv) neoangiogenesis is altered with increased arterial damage. MATERIALS: Thirty-three patients with PAOD and critical lower limb ischaemia, 22 with type II diabetes. METHODS: Immunohistochemistry for endothelial cell markers (CD34 and von Willebrand Factor); real-time reverse transcription polymerase chain reaction (RT-PCR) to quantify arterial wall expression of vascular endothelial growth factor (VEGF); enzyme-linked immunosorbent assay (ELISA) to assess blood VEGF; flow cytometry to detect circulating endothelial cells (CECs). RESULTS: Patients with PAOD and diabetes have a higher frequency (60% vs. 45%) of advanced atherosclerotic lesions and a significant reduction (p = 0.0003) in CD34(+) capillaries in the arterial media. Adventitial neoangiogenesis was increased equally (CD34(+) and vWF(+)) in all patients. Likewise, all patients have increased CEC and VEGF concentration in the blood as well as in-situ VEGF transcript expression. CONCLUSIONS: Patients with PAOD have remarkable arterial damage despite increased in-situ and circulating expression of the pro-angiogenic VEGF; a dysfunctional vv angiogenesis was seen in diabetics which also showed a higher frequency of parietal damage; it is suggested that in diabetic arterial wall, injury is worsened by vv inability to finalise an effective VEGF-driven arterial wall neoangiogenesis.


Subject(s)
Arterial Occlusive Diseases/physiopathology , Diabetic Angiopathies/physiopathology , Ischemia/physiopathology , Lower Extremity/blood supply , Lower Extremity/physiopathology , Neovascularization, Physiologic , Vasa Vasorum/physiopathology , Adult , Aged , Aged, 80 and over , Antigens, CD34/analysis , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/pathology , Case-Control Studies , Critical Illness , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Endothelial Cells/pathology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immunohistochemistry , Ischemia/metabolism , Ischemia/pathology , Italy , Male , Middle Aged , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Vasa Vasorum/chemistry , Vasa Vasorum/pathology , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/genetics , von Willebrand Factor/analysis
12.
Leukemia ; 24(4): 687-98, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20200557

ABSTRACT

Alkylphospholipids and alkylphosphocholines (APCs) are promising antitumor agents, which target the plasma membrane and affect multiple signal transduction networks. We investigated the therapeutic potential of erucylphosphohomocholine (ErPC3), the first intravenously applicable APC, in human acute myelogenous leukemia (AML) cells. ErPC3 was tested on AML cell lines, as well as AML primary cells. At short (6-12 h) incubation times, the drug blocked cells in G2/M phase of the cell cycle, whereas, at longer incubation times, it decreased survival and induced cell death by apoptosis. ErPC3 caused JNK 1/2 activation as well as ERK 1/2 dephosphorylation. Pharmacological inhibition of caspase-3 or a JNK 1/2 inhibitor peptide markedly reduced ErPC3 cytotoxicity. Protein phosphatase 2A downregulation by siRNA opposed ERK 1/2 dephosphorylation and blunted the cytotoxic effect of ErPC3. ErPC3 was cytotoxic to AML primary cells and reduced the clonogenic activity of CD34(+) leukemic cells. ErPC3 induced a significant apoptosis in the compartment (CD34(+) CD38(Low/Neg) CD123(+)) enriched in putative leukemia-initiating cells. This conclusion was supported by ErPC3 cytotoxicity on AML blasts showing high aldehyde dehydrogenase activity and on the side population of AML cell lines and blasts. These findings indicate that ErPC3 might be a promising therapeutic agent for the treatment of AML patients.


Subject(s)
Apoptosis/drug effects , Erucic Acids/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Kinase 4/metabolism , Phosphorylcholine/analogs & derivatives , Precursor Cells, B-Lymphoid/drug effects , Protein Phosphatase 2/metabolism , Animals , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , 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 , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylcholine/pharmacology , Precursor Cells, B-Lymphoid/metabolism , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
13.
Int J Immunopathol Pharmacol ; 22(3): 699-706, 2009.
Article in English | MEDLINE | ID: mdl-19822086

ABSTRACT

Dental pulp is a heterogeneous microenviroment where unipotent progenitor and pluripotent mesenchymal stem cells cohabit. In this study we investigated whether human dental pulp stromal (stem) cells (DP-SCs) committed to the angiogenic fate. DP-SCs showed the specific mesenchymal immunophenotypical profile positive for CD29, CD44, CD73, CD105, CD166 and negative for CD14, CD34, CD45, in accordance with that reported for bone marrow-derived SCs. The Oct-4 expression in DP-SCs, evaluated through RT-PCR analysis, increased in relation with the number of the passages in cell culture and decreased after angiogenic induction. In agreement with their multipotency, DP-SCs differentiated toward osteogenic and adipogenic commitments. In angiogenic experiments, differentiation of DP-SCs, through vascular endothelial growth factor (VEGF) induction, was evaluated by in vitro matrigel assay and by cytometric analysis. Accordingly, endothelial-specific markers like Flt-1 and KDR were basally expressed and they increased after exposure to VEGF together with the occurrence of ICAM-1 and von Willebrand factor positive cells. In addition, VEGF-induced DP-SCs maintained endothelial cell-like features when cultured in a 3-D fibrin mesh, displaying focal organization into capillary-like structures. The DP-SC angiogenic potential may prove a remarkable tool for novel approaches to developing tissue-engineered vascular grafts which are useful when vascularization of ischemic tissues is required.


Subject(s)
Adult Stem Cells/physiology , Dental Pulp/physiology , Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Neovascularization, Physiologic , Stromal Cells/physiology , Tissue Engineering , Adult , Adult Stem Cells/immunology , Adult Stem Cells/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Separation , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/immunology , Dental Pulp/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Fibrin/metabolism , Flow Cytometry , Humans , Immunophenotyping , Intercellular Adhesion Molecule-1/metabolism , Male , Microscopy, Electron, Transmission , Octamer Transcription Factor-3/genetics , RNA, Messenger/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Stromal Cells/immunology , Stromal Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , von Willebrand Factor/metabolism
14.
Leukemia ; 22(1): 147-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17928881

ABSTRACT

The serine/threonine kinase Akt, a downstream effector of phosphatidylinositol 3-kinase (PI3K), is known to play an important role in antiapoptotic signaling and has been implicated in the aggressiveness of a number of different human cancers including acute myelogenous leukemia (AML). We have investigated the therapeutic potential of the novel Akt inhibitor, perifosine, on human AML cells. Perifosine is a synthetic alkylphospholipid, a new class of antitumor agents, which target plasma membrane and inhibit signal transduction networks. Perifosine was tested on THP-1 and MV 4-11 cell lines, as well as primary leukemia cells. Perifosine treatment induced cell death by apoptosis in AML cell lines. Perifosine caused Akt and ERK 1/2 dephosphorylation as well as caspase activation. In THP-1 cells, the proapoptotic effect of perifosine was partly dependent on the Fas/FasL system and c-jun-N-kinase activation. In MV 4-11 cells, perifosine downregulated phosphorylated Akt, but not phosphorylated FLT3. Moreover, perifosine reduced the clonogenic activity of AML, but not normal, CD34(+) cells, and markedly increased blast cell sensitivity to etoposide. Our findings indicate that perifosine, either alone or in combination with existing drugs, might be a promising therapeutic agent for the treatment of those AML cases characterized by upregulation of the PI3K-Akt survival pathway.


Subject(s)
Apoptosis/drug effects , Leukemia, Myeloid, Acute/drug therapy , Phosphorylcholine/analogs & derivatives , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Blotting, Western , Cell Proliferation/drug effects , Colony-Forming Units Assay , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Leukemia, Myeloid, Acute/metabolism , MAP Kinase Kinase 4/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylcholine/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , bcl-Associated Death Protein/metabolism , fas Receptor/metabolism , fms-Like Tyrosine Kinase 3/metabolism
15.
Curr Med Chem ; 14(19): 2009-23, 2007.
Article in English | MEDLINE | ID: mdl-17691943

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K)/Akt (protein kinase B, PKB)/mammalian Target Of Rapamycin (mTOR) signaling pathway plays a critical role in many cellular functions which are elicited by extracellular stimuli. However, constitutively active PI3K/Akt/mTOR signaling has also been firmly established as a major determinant for cell growth, proliferation, and survival in an wide array of human cancers. Thus, blocking the PI3K/AKT/mTOR signal transduction network could be an effective new strategy for targeted anticancer therapy. Pharmacological inhibitors of this signaling cascade are powerful antineoplastic agents in vitro and in xenografted models of tumors, and some of them are now being tested in clinical trials. Recent studies showed that PI3K/Akt/mTOR axis is frequently activated in acute myelogenous leukemia (AML) patient blasts and strongly contributes to proliferation, survival, and drug-resistance of these cells. Both the disease-free survival and overall survival are significantly shorter in AML cases with PI3K/Akt/mTOR upregulation. Therefore, this signal transduction cascade may represent a target for innovative therapeutic treatments of AML patients. In this review, we discuss the possible mechanisms of activation of this pathway in AML cells and the downstream molecular targets of the PI3K/Akt/mTOR signaling network which are important for blocking apoptosis, enhancing proliferation, and promoting drug-resistance of leukemic cells. We also highlight several pharmacological inhibitors which have been used to block this pathway for targeted therapy of AML. These small molecules induce apoptosis or sensitize AML cells to existing drugs, and might be used in the future for improving the outcome of this hematological disorder.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Antibiotics, Antineoplastic/therapeutic use , Apoptosis , Cell Cycle , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
16.
Leukemia ; 21(5): 886-96, 2007 May.
Article in English | MEDLINE | ID: mdl-17361225

ABSTRACT

Insulin-like growth factor-I (IGF-I) and its receptor (IGF-IR) have been implicated in the pathophysiology of many human cancers, including those of hematopoietic lineage. We investigated the therapeutic potential of the novel IGF-IR tyrosine kinase activity inhibitor, NVP-AEW541, on human acute myeloid leukemia (AML) cells. NVP-AEW541 was tested on a HL60 cell subclone, which is dependent on autocrine secretion of IGF-I for survival and drug resistance, as well as primary drug resistant leukemia cells. NVP-AEW541 treatment (24 h) induced dephosphorylation of IGF-IR. NVP-AEW541 also caused Akt dephosphorylation and changes in the expression of key regulatory proteins of the cell cycle. At longer incubation times (48 h), NVP-AEW541-induced apoptotic cell death, as demonstrated by caspase-3 cleavage. Apoptosis was accompanied by decreased expression of anti-apoptotic proteins. NVP-AEW541 enhanced sensitivity of HL60 cells to either cytarabine or etoposide. Moreover, NVP-AEW541 reduced the clonogenic capacity of AML CD34(+) cells cultured in the presence of IGF-I. Chemoresistant AML blasts displayed enhanced IGF-I secretion, and were sensitized to etoposide-induced apoptosis by NVP-AEW541. Our findings indicate that NVP-AEW541 might be a promising therapeutic agent for the treatment of those AML cases characterized by IGF-I autocrine secretion.


Subject(s)
Apoptosis/drug effects , Insulin-Like Growth Factor I/metabolism , Leukemia, Myeloid, Acute/drug therapy , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p27 , Cytarabine/pharmacology , Down-Regulation , Etoposide/pharmacology , HL-60 Cells , Humans , Intracellular Signaling Peptides and Proteins/analysis , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Receptor, IGF Type 1/metabolism
17.
Leukemia ; 21(3): 427-38, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17215852

ABSTRACT

A high incidence of relapses following induction chemotherapy is a major hindrance to patient survival in acute myelogenous leukemia (AML). There is strong evidence that activation of the phosphoinositide 3 kinase (PI3K)/Akt signaling network plays a significant role in rendering AML blasts drug resistant. An important mechanism underlying drug resistance is represented by overexpression of membrane drug transporters such as multidrug resistance-associated protein 1 (MRP1) or 170-kDa P-glycoprotein (P-gp). Here, we present evidence that MRP1, but not P-gp, expression is under the control of the PI3K/Akt axis in AML blasts. We observed a highly significant correlation between levels of phosphorylated Akt and MRP1 expression in AML cells. Furthermore, incubation of AML blasts with wortmannin, a PI3K pharmacological inhibitor, resulted in lower levels of phosphorylated Akt, downregulated MRP1 expression, and decreased Rhodamine 123 extrusion in an in vitro functional dye efflux assay. We also demonstrate that wortmannin-dependent PI3K/Akt inhibition upregulated p53 protein levels in most AML cases, and this correlated with diminished MRP1 expression and enhanced phosphorylation of murine double minute 2 (MDM2). Taken together, these data suggest that PI3K/Akt activation may lead to the development of chemoresistance in AML blasts through a mechanism involving a p53-dependent suppression of MRP1 expression.


Subject(s)
Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Leukemic/physiology , Leukemia, Myeloid/pathology , Multidrug Resistance-Associated Proteins/biosynthesis , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acute Disease , Adult , Aged , Aged, 80 and over , Androstadienes/pharmacology , Bone Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Female , Fluorescent Dyes/metabolism , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Genes, p53 , Humans , Jurkat Cells/drug effects , Jurkat Cells/metabolism , Leukemia, Myeloid/genetics , Leukemia, Myeloid/metabolism , Leukemia, Promyelocytic, Acute/pathology , Leukemia-Lymphoma, Adult T-Cell/pathology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Osteosarcoma/pathology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , Rhodamine 123/metabolism , Tumor Suppressor Protein p53/biosynthesis , Wortmannin
18.
Cell Tissue Bank ; 8(3): 195-203, 2007.
Article in English | MEDLINE | ID: mdl-17063259

ABSTRACT

This study was aimed to establish whether the cryopreservation procedure we currently use in clinics can modify arterial homograft antigenicity. To this purpose, we performed an immunohistochemical study on fresh and cryopreserved human arterial homografts to visualize the expression of HLA class I heavy and light chains "in situ" by using the HC-10 and Namb-1 monoclonal antibodies. Human femoral arteries and thoracic aortas were harvested from 18 heart-beating donors and sampled before and after cryopreservation. Arterial segments were frozen in liquid nitrogen vapors in a controlled rate freezing system. After thawing, samples were processed for routine immunohistochemistry. To standardize immunostaining, flow-cytometry indirect immunofluorescence analysis was performed on HUVEC; immunohistochemistry of human ovarian cortical vessels was performed as an additional positive control. Negative controls were performed by omitting tissue incubation with primary antibodies. HLA-class I antigens were markedly expressed by endothelial cells lining surface intima and adventitial vasa vasorum; a moderate expression was found in medial smooth muscle cells. Except for the surface unreactivity caused by loss of endothelium, results from cryopreserved arterial allografts were strictly comparable to those observed in fresh, unfrozen tissues. These results support the view that cryopreserved arterial allografts are immunogenic as their fresh counterparts; apart from smooth muscle cells which retained a moderate expression of HLA class I antigens following cryopreservation, our study suggests that the highly HC-10 positive endothelial cells we found to line the rich adventitial network of vasa vasorum are expected to be one of the major targets of the serological response in the recipient.


Subject(s)
Arteries/immunology , Arteries/transplantation , Cryopreservation , HLA Antigens/immunology , Adult , Antibodies, Monoclonal/immunology , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Transplantation, Homologous
19.
Leukemia ; 20(6): 911-28, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16642045

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/Akt signaling pathway is crucial to many aspects of cell growth, survival and apoptosis, and its constitutive activation has been implicated in the both the pathogenesis and the progression of a wide variety of neoplasias. Hence, this pathway is an attractive target for the development of novel anticancer strategies. Recent studies showed that PI3K/Akt signaling is frequently activated in acute myeloid leukemia (AML) patient blasts and strongly contributes to proliferation, survival and drug resistance of these cells. Upregulation of the PI3K/Akt network in AML may be due to several reasons, including FLT3, Ras or c-Kit mutations. Small molecules designed to selectively target key components of this signal transduction cascade induce apoptosis and/or markedly increase conventional drug sensitivity of AML blasts in vitro. Thus, inhibitory molecules are currently being developed for clinical use either as single agents or in combination with conventional therapies. However, the PI3K/Akt pathway is important for many physiological cellular functions and, in particular, for insulin signaling, so that its blockade in vivo might cause severe systemic side effects. In this review, we summarize the existing knowledge about PI3K/Akt signaling in AML cells and we examine the rationale for targeting this fundamental signal transduction network by means of selective pharmacological inhibitors.


Subject(s)
Leukemia, Myeloid/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Acute Disease , Humans , Leukemia, Myeloid/drug therapy , Models, Biological , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects
20.
Cryobiology ; 52(2): 309-16, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16458877

ABSTRACT

The cryopreservation protocol we use for arterial reconstructive surgery has been studied to evaluate smooth muscle cell (SMC) structural integrity and viability before implantation. Samples of human thoracic aortas (HTA) were harvested from five multi-organ donors. Sampling included unfrozen and cryopreserved specimens. Cryopreservation was performed using RPMI with human albumin and 10% Me(2)SO in a controlled-rate freezing apparatus. Thawing was accomplished by submerging bags in a water bath (39 degrees C) followed by washings in cooled saline. In situ cell preservation as investigated by light and transmission electron microscopy showed that SMCs from cryopreserved HTA had nuclear and cytoplasmic changes. A TUNEL assay, performed to detect DNA fragmentation in situ, showed increased SMC nuclear positivity in cryopreserved HTA when compared to unfrozen samples. 7-AAD flow cytometry assay of cells derived from cryopreserved HTA showed that an average of 49+/-16% cells were unlabeled after cryopreservation. Organ cultures aimed to study cell ability to recover cryopreservation damage showed a decreasing number of SMCs from day 4 to day 15 in cryopreserved HTA. In conclusion, the cryopreservation protocol applied in this study induces irreversible damage of a significant fraction of arterial SMCs.


Subject(s)
Aorta, Thoracic , Cryopreservation , Myocytes, Smooth Muscle/pathology , Organ Preservation , Tissue Survival , Aorta, Thoracic/injuries , Cryoprotective Agents/chemistry , Humans , In Situ Nick-End Labeling , Microscopy, Electron, Transmission , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...