Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Transl Oncol ; 11(2): 406-415, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29459325

ABSTRACT

Antiangiogenics administration in colorectal cancer patients seemed promising therapeutic approach. Inspite of early encouraging results, it however gave only modest clinical benefits. When AAG was administered with discontinuous schedule, the disease showed acceleration in certain cases. Though resistance to AAG has been extensively studied, it is not documented for discontinuous schedules. To simulate clinical situations, we subjected a patient-derived CRC subcutaneous xenograft in mice to three different protocols: 1) AAG (bevacizumab) treatment for 30 days (group A) (group B was the control), 2) bevacizumab treatment for 50 days (group C) and bevacizumab for 30 days and 20 without treatment (group D), and 3) bevacizumab treatment for 70 days (group E) and 70 days treatment with a drug-break period between day 30 and 50 (group F). The tumor growth was monitored, and at sacrifice, the vascularity of tumors was measured and the proangiogenic factors quantified. Tumor phenotype was studied by quantifying cancer stem cells. Interrupting bevacizumab during treatment accelerated tumor growth and revascularization. A significant increase of proangiogenic factors was observed when therapy was stopped. On withdrawal of bevacizumab, as also after the drug-break period, the plasmatic VEGF increased significantly. Similarly, a notable increase of CSCs after the withdrawal and drug-break period of bevacizumab was observed (P<.01). The present study indicates that bevacizumab treatment needs to be maintained because discontinuous schedules tend to trigger tumor regrowth, and increase tumor resistance and CSC heterogeneity.

2.
Curr Alzheimer Res ; 15(7): 643-654, 2018.
Article in English | MEDLINE | ID: mdl-29357794

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) and age-related macular degeneration (AMD) present similarities, particularly with respect to oxidative stress, including production of 4-Hydroxy-2- nonenal (HNE). AMD has been named the AD in the eye. The Müller cells (MC) function as a principal glia of the retina and maintain water/potassium, glutamate homeostasis and redox status. Any MC dysfunction results in retinal neurodegeneration. OBJECTIVES: We investigated the effects of HNE in human MC. RESULTS: HNE induced an increase of the reactive oxygen species associated with mitochondrial dysfunction and apoptosis. HNE induced endoplasmic reticulum (ER) stress (upregulation of GRP78/Bip, and the proapoptotic factor, CHOP). HNE also impaired expression of genes controlling potassium homeostasis (KCNJ10), glutamate detoxification (GS), and the visual cycle (RLBP1). MC adaptive response to HNE included upregulation of amyloid-ß protein precursor (AßPP). To determine the role of AßPP, we overexpressed AßPP in MC. Overexpression of AßPP induced strong antioxidant and anti-ER stress (PERK downregulation and GADD34 upregulation) responses accompanied by activation of the prosurvival branch of the unfolded protein response. It was also associated with upregulation of major genes involved in MC-controlled retinal homeostasis (KCNJ10, GS, and RLBP1) and protection against HNE-induced apoptosis. Therefore, AßPP is an ER and oxidative stress responsive molecule, and is able to stimulate the transcription of major genes involved in MC functions impaired by HNE. CONCLUSION: Our study suggests that targeting oxidative and ER stress might be a potential therapeutic strategy against glia impairment in AMD and AD, in light of the common features between the two pathologies.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Cell Survival/physiology , Neuroglia/metabolism , Oxidative Stress/physiology , Transcriptome , Unfolded Protein Response/physiology , Amyloid beta-Protein Precursor/genetics , Cell Death/physiology , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum Chaperone BiP , Humans , Mitochondria/metabolism , Neuroprotection/physiology , Reactive Oxygen Species/metabolism , Transcription, Genetic/physiology
3.
Stem Cells ; 33(8): 2431-41, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25850942

ABSTRACT

While enucleation is a critical step in the terminal differentiation of human red blood cells, the molecular mechanisms underlying this unique process remain unclear. To investigate erythroblast enucleation, we studied the erythroid differentiation of human embryonic stem cells (hESCs), which provide a unique model for deeper understanding of the development and differentiation of multiple cell types. First, using a two-step protocol, we demonstrated that terminal erythroid differentiation from hESCs is directly dependent on the age of the embryoid bodies. Second, by choosing hESCs in two extreme conditions of erythroid culture, we obtained an original differentiation model which allows one to study the mechanisms underlying the enucleation of erythroid cells by analyzing the gene and miRNA (miR) expression profiles of cells from these two culture conditions. Third, using an integrated analysis of mRNA and miR expression profiles, we identified five miRs potentially involved in erythroblast enucleation. Finally, by selective knockdown of these five miRs we found miR-30a to be a regulator of erythroblast enucleation in hESCs.


Subject(s)
Cell Differentiation , Erythroblasts/metabolism , Gene Expression Regulation , Human Embryonic Stem Cells/metabolism , MicroRNAs/biosynthesis , RNA, Messenger/biosynthesis , Cells, Cultured , Erythroblasts/cytology , Human Embryonic Stem Cells/cytology , Humans
4.
Dis Model Mech ; 7(11): 1253-61, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25261568

ABSTRACT

In the translational process of developing innovative therapies for DMD (Duchenne muscular dystrophy), the last preclinical validation step is often carried out in the most relevant animal model of this human disease, namely the GRMD (Golden Retriever muscular dystrophy) dog. The disease in GRMD dogs mimics human DMD in many aspects, including the inter-individual heterogeneity. This last point can be seen as a drawback for an animal model but is inherently related to the disease in GRMD dogs closely resembling that of individuals with DMD. In order to improve the management of this inter-individual heterogeneity, we have screened a combination of biomarkers in sixty-one 2-month-old GRMD dogs at the onset of the disease and a posteriori we addressed their predictive value on the severity of the disease. Three non-invasive biomarkers obtained at early stages of the disease were found to be highly predictive for the loss of ambulation before 6 months of age. An elevation in the number of circulating CD4(+)CD49d(hi) T cells and a decreased stride frequency resulting in a reduced spontaneous speed were found to be strongly associated with the severe clinical form of the disease. These factors can be used as predictive tests to screen dogs to separate them into groups with slow or fast disease progression before their inclusion into a therapeutic preclinical trial, and therefore improve the reliability and translational value of the trials carried out on this invaluable large animal model. These same biomarkers have also been described to be predictive for the time to loss of ambulation in boys with DMD, strengthening the relevance of GRMD dogs as preclinical models of this devastating muscle disease.


Subject(s)
Biomarkers/blood , Disease Models, Animal , Muscular Dystrophy, Duchenne/physiopathology , Animals , Dogs , Immunophenotyping , Muscular Dystrophy, Duchenne/immunology
5.
Int J Oncol ; 45(4): 1372-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25095896

ABSTRACT

The importance of the insulin-like growth factor, IGF, as a signaling axis in cancer development, progression and metastasis is highlighted by its effects on cancer cells, notably proliferation and acquired resistance. The role of the microenvironment within which cancer cells evolve and which mediates this effect is far from clear. Here, the involvement of IGF-I in inducing multidrug resistance in a myeloid leukemia cell line, grown in the presence of bone marrow-derived stromal cells called 'Hospicells' (BMH), is demonstrated. We found that i) drug sensitive as well as resistant leukemia cells express IGF-I and its receptor IGF-IR. However, the resistant cells were found to secrete high levels of IGF-I. ii) Presence of exogenous IGF-I promoted cell proliferation, which decreased when an inhibitor of IGF-IR (picropodophyllin, PPP) was added. iii) BMH and IGF-I are both involved in the regulation of genes of the ATP binding cassette (ABC) related to resistance development (MDR1, MRP1, MRP2, MRP3 and BCRP). iv) The levels of ABC gene expression by leukemia cells were found to increase in the presence of increasing numbers of BMH. However, these levels decreased when IGF-IR was inhibited by addition of PPP. v) Co-culture of the drug-sensitive leukemia cells with BMH induced protection against the action of daunorubicin. This chemoresistance was amplified by the presence of IGF-I whereas it decreased when IGF-IR was inhibited. Our results underline the role of microenvironment in concert with the IGF-1 pathway in conferring drug resistance to leukemia cells.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Insulin-Like Growth Factor I/pharmacology , Leukemia/pathology , Mesenchymal Stem Cells/metabolism , Podophyllotoxin/analogs & derivatives , ATP-Binding Cassette Transporters/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Daunorubicin/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Leukemia/drug therapy , Leukemia/metabolism , Podophyllotoxin/pharmacology , Receptor, IGF Type 1 , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism
6.
Int J Oncol ; 43(4): 1011-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23877403

ABSTRACT

In spite of the growing importance of endothelial protein C receptor/active protein C (EPCR/aPC) in tumor biology, their impact on immunological homeostasis remains largely unexplored. The objective of this study was to assess whether soluble plasma endothelial protein C receptor (sEPCR), which is a regulator of circulating aPC, is involved in innate immune response in cancer patients. In the Ovcar-3 ovarian cancer line, the role of aPC in secretion of cytokines was analyzed. In parallel, in 33 patients, with a diagnosis of ovarian epithelial cancer, sEPCR was quantified, blood immune cell phenotypes were determined by flow cytometry and plasma cytokines were evaluated using a protein array. Spearman's rank correlation coefficients (r) and coefficient significance was determined by a statistical hypothesis test (α=0.05). Our results show that i) aPC induced the secretion of several cytokines in Ovcar-3 cells; ii) 61% of patients exhibited a concentration of plasma sEPCR well above the baseline (normal plasma level, 100 ± 28 ng/ml); iii) comparing immune cell phenotypes in patients having a normal level of sEPCR with those having a high level of sEPCR, it was found that sEPCR levels were correlated with high intensity of cells expressing CD45ra, CD3, CD8, CD25 and low intensity of cells expressing CD56 (NK cells), CD294 (TH2 cells), IL-2, IL-10, IL-17a (TH17 cells), IL-21 (TH21 cells) and CD29 markers (r ≥ 0.60); and iv) high levels of sEPCR correlate with high levels of plasma bioactive proteins such as insulin-like growth factor-2 (IGFII), IL-13rα, macrophage inflammatory protein (MIP1α) and matrix metalloproteinase-7 (MMP-7) that have already been proposed as biomarkers for ovarian cancer and particularly those with poor prognosis. In conclusion, sEPCR produced by ovarian cancer cells, by modulating circulating aPC, influences the secretory behavior of tumor cells (cytokines and interleukins). Consequently, sEPCR in turn acts on the innate immune response by decreasing effector cells such as natural killer and T helper cells (TH2, TH17 and TH21).


Subject(s)
Antigens, CD/biosynthesis , Immunity, Innate/genetics , Killer Cells, Natural/immunology , Ovarian Neoplasms/immunology , Protein C/biosynthesis , Receptors, Cell Surface/biosynthesis , Th17 Cells/immunology , Adult , Antigens, CD/blood , Antigens, CD/genetics , Cell Line, Tumor , Endothelial Protein C Receptor , Female , Flow Cytometry , Humans , Interleukin-10/blood , Interleukin-2/blood , Interleukins/blood , Killer Cells, Natural/pathology , Matrix Metalloproteinase 7/blood , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein C/metabolism , Receptors, Cell Surface/blood , Receptors, Cell Surface/genetics , Th17 Cells/pathology
7.
J Surg Res ; 184(2): 888-97, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23726239

ABSTRACT

BACKGROUND: Hepatic pedicle clamping is often required to reduce blood loss and transfusion during liver resection. However, the question remains whether use of hepatic pedicle clamping promotes tumor growth. Endothelial progenitor cells (EPCs) are mobilized from bone marrow in response to tissue ischemia, which allows neovascularization of ischemic tissue. It has been suggested that EPCs are involved in tumor progression. We hypothesized that hepatic ischemia reperfusion (I/R)-induced mobilization of EPCs could enhance growth of microscopic tumor, therefore promoting liver metastasis in a mouse model of colorectal cancer. MATERIALS AND METHODS: We used mouse models of hepatic I/R and hind limb ischemia. For comparison, we studied mice that underwent limb ischemia as positive controls of EPC mobilization. At day 0, we divided 40 mice into four groups: hepatic I/R, hind limb ischemia, combined hepatic I/R and hind limb ischemia, and control (sham midline incision laparotomy). At day 2, we induced liver metastasis in all mice by injecting CT-26 cells into the spleen. Time-dependent circulating EPCs were determined by flow cytometry. We evaluated liver metastasis and microvascular density on day 21. RESULTS: The number of circulating progenitor cells increased rapidly in the ischemic groups compared with the control group. Hepatic I/R significantly increased tumor outgrowth compared with the control group. Increased tumor growth was associated with enhanced CD31-positive microvascular density in liver tissue. CONCLUSIONS: Hepatic I/R leads to mobilization of bone marrow-derived EPCs and enhanced intra-hepatic angiogenesis, which is associated with increased tumor burden in an animal model of colorectal liver metastasis.


Subject(s)
Bone Marrow Cells/pathology , Cell Proliferation , Colorectal Neoplasms/pathology , Hematopoietic Stem Cells/pathology , Liver Neoplasms/secondary , Liver/blood supply , Reperfusion Injury/physiopathology , Animals , Cell Count , Cell Line, Tumor , Chemokine CXCL12/blood , Disease Models, Animal , Disease Progression , Female , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic/physiopathology
8.
PLoS One ; 8(5): e62303, 2013.
Article in English | MEDLINE | ID: mdl-23658722

ABSTRACT

A role for caspase activation in erythroid differentiation has been established, yet its precise mode of action remains elusive. A drawback of all previous investigations on caspase activation in ex vivo erythroid differentiation is the lack of an in vitro model producing full enucleation of erythroid cells. Using a culture system which renders nearly 100% enucleated red cells from human CD34(+) cells, we investigated the role of active caspase-3 in erythropoiesis. Profound effects of caspase-3 inhibition were found on erythroid cell growth and differentiation when inhibitors were added to CD34(+) cells at the start of the culture and showed dose-response to the concentration of inhibitor employed. Enucleation was only reduced as a function of the reduced maturity of the culture and the increased cell death of mature cells while the majority of cells retained their ability to extrude their nuclei. Cell cycle analysis after caspase-3 inhibition showed caspase-3 to play a critical role in cell proliferation and highlighted a novel function of this protease in erythroid differentiation, i.e. its contribution to cell cycle regulation at the mitotic phase. While the effect of caspase-3 inhibitor treatment on CD34(+) derived cells was not specific to the erythroid lineage, showing a similar reduction of cell expansion in myeloid cultures, the mechanism of action in both lineages appeared to be distinct with a strong induction of apoptosis causing the decreased yield of myeloid cells. Using a series of colony-forming assays we were able to pinpoint the stage at which cells were most sensitive to caspase-3 inhibition and found activated caspase-3 to play a signalling role in erythroid differentiation by targeting mature BFU-E and CFU-E but not early BFU-E.


Subject(s)
Caspase 3/metabolism , Cell Differentiation , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/enzymology , Signal Transduction , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Erythroid Precursor Cells/drug effects , G2 Phase/drug effects , Humans , Signal Transduction/drug effects
9.
Cell Mol Life Sci ; 70(4): 729-42, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23007843

ABSTRACT

Links between cancer and stem cells have been proposed for many years. As the cancer stem cell (CSC) theory became widely studied, new methods were developed to culture and expand cancer cells with conserved determinants of "stemness". These cells show increased ability to grow in suspension as spheres in serum-free medium supplemented with growth factors and chemicals. The physiological relevance of this phenomenon in established cancer cell lines remains unclear. Cell lines have traditionally been used to explore tumor biology and serve as preclinical models for the screening of potential therapeutic agents. Here, we grew cell-forming spheres (CFS) from 25 established colorectal cancer cell lines. The molecular and cellular characteristics of CFS were compared to the bulk of tumor cells. CFS could be isolated from 72 % of the cell lines. Both CFS and their parental CRC cell lines were highly tumorigenic. Compared to their parental cells, they showed similar expression of putative CSC markers. The ability of CRC cells to grow as CFS was greatly enhanced by prior treatment with 5-fluorouracil. At the molecular level, CFS and parental CRC cells showed identical gene mutations and very similar genomic profiles, although microarray analysis revealed changes in CFS gene expression that were independent of DNA copy-number. We identified a CFS gene expression signature common to CFS from all CRC cell lines, which was predictive of disease relapse in CRC patients. In conclusion, CFS models derived from CRC cell lines possess interesting phenotypic features that may have clinical relevance for drug resistance and disease relapse.


Subject(s)
Colorectal Neoplasms/pathology , Spheroids, Cellular/pathology , Animals , Antimetabolites, Antineoplastic/pharmacology , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/drug effects , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Fluorouracil/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Recurrence, Local , Rectum/drug effects , Rectum/metabolism , Rectum/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Tumor Cells, Cultured
10.
Eur J Pharmacol ; 685(1-3): 1-7, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22542663

ABSTRACT

We have previously demonstrated that matrix metalloproteinase-9 (MMP-9) is critical for breast cancer cell migration and is necessary but not sufficient for tubular network formation. Given the important angiogenic activity of vascular endothelial growth factor (VEGF), we investigate here its possible contribution in tubular network formation and its link with MMP-9. Exposure of resistant epithelial breast cancer cells (rMCF-7) to Avastin, a VEGF neutralising antibody, suppresses tubular network formation but not cell migration. However, their exposure to MMP-9 inhibitor markedly decreases both parameters. Besides, the addition of exogenous VEGF or MMP-9 alone or in combination to sensitive parental cells (sMCF-7) or rMCF-7 cells enhances tubular network formation by rMCF-7 cells but not by sMCF-7 cells. The evaluation of the expression levels of VEGF receptor (VEGFR) subtypes shows that sMCF-7 cells express only small quantities of VEGFR-2 and VEGFR-3 compared with rMCF-7 cells that express strong quantities. However, treatment of sMCF-7 cells by phorbol 12-myristate 13-acetate (PMA), a PKC activator, induces both tubular network formation and VEGFR-2/VEGFR-3 over-expressions. Interestingly, exposure of rMCF-7 cells or PMA-treated sMCF-7 cells to the specific inhibitors of VEGFR-2 and VEGFR-3 reduces markedly the tubular network formation. Together, our results demonstrate that the proteolytic enzyme MMP-9 promotes rMCF-7 cell migration and, consequently, tubular network formation through VEGFR-2/ VEGFR-3 activation. Understanding of mechanisms involved in vasculogenic mimicry and cell migration related to MMP-9 and VEGF may open new opportunities to improve cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Matrix Metalloproteinase 9/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Antibiotics, Antineoplastic/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Bevacizumab , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 9/administration & dosage , Tetradecanoylphorbol Acetate/pharmacology , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
11.
Breast Cancer Res Treat ; 131(1): 49-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21336598

ABSTRACT

The purpose of this article is to determine the tumorigenic potential of estradiol treatment (E2) when combined with either progesterone (P4) or medroxyprogesterone acetate (MPA) in normal luminal human breast cells (HBE) and in human breast cancer cells (T47-D, MCF-7). Proliferation profiles were evaluated, along with the gene transactivation activity between the progesterone and glucocorticoid receptors (PR, GR) in HBE, T47-D, and MCF-7 cells treated by E2 + P4 or E2 + MPA. High throughput transcriptome analysis was performed on RNA from HBE cells treated by E2, E2 + MPA and E2 + P4. GR content was analyzed in normal breast cells as well. In HBE cells, E2 + P4 treatment was antiproliferative and promoted cellular differentiation. In contrast, E2 + MPA displayed mitogenic, antiapoptotic effects in HBE cells and did not influence cellular differentiation. The effect of P4 and MPA on cell proliferation was, however, variable in breast cancer cells. In cells containing GR or/and PR, MPA decreased proliferation whereas P4 antiproliferative effect needed the presence of PR. In HBE cells, the regulation of genes by E2 + P4, and E2 + MPA was significantly different, particularly in cell proliferation and cell death gene families. Further analysis revealed a modulation of the glucocorticoid receptor gene expression pathway by E2 + MPA. Predominant MPA glucocorticoid activity in normal and breast cancer cells was demonstrated using a glucocorticoid antagonist and the down-regulation of the GR by RNA interference. In normal luminal breast cells and in breast cancer cells, P4 and MPA combined with E2 treatment have opposing mitogenic effects due to GR. The consequences of MPA glucocorticoid potencies as well as the importance of GR in breast tissue merit a reappraisal.


Subject(s)
Estradiol/pharmacology , Medroxyprogesterone Acetate/pharmacology , Progesterone/pharmacology , Receptors, Glucocorticoid/metabolism , Receptors, Progesterone/metabolism , Adolescent , Adult , Breast , Breast Neoplasms , Carrier Proteins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Nucleotide Sequencing , Hormone Replacement Therapy/methods , Humans , Norpregnadienes/pharmacology , RNA Interference , RNA, Small Interfering , Receptors, Estrogen/metabolism , Steroids , Transcriptome , Young Adult
12.
Cancer Med ; 1(2): 261-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23342274

ABSTRACT

Elevated plasma level of soluble endothelial protein C receptor (sEPCR) may be an indicator of thrombotic risk. The present study aims to correlate leukemia-associated hypercoagulability to high level plasma sEPCR and proposes its measurement in routine clinical practice. EPCR expressions in leukemic cell lines were determined by flow cytometry, immunocytochemistry, and reverse transcription polymerase chain reaction (RT-PCR). EPCR gene sequence of a candidate cell line HL-60 was also determined. Plasma samples (n = 76) and bone marrow aspirates (n = 72) from 148 patients with hematologic malignancies and 101 healthy volunteers were analyzed by enzyme-linked immunosorbent assay (ELISA) via a retrospective study for sEPCR and D-dimer. All leukemic cell lines were found to express EPCR. Also, HL-60 EPCR gene sequence showed extensive similarities with the endothelial reference gene. All single nucleotide polymorphisms (SNPs) originally described and some new SNPs were revealed in the promoter and intronic regions. Among these patients 67% had plasma sEPCR level higher than the controls (100 ± 28 ng/mL), wherein 16.3% patients had experienced a previous thrombotic event. These patients were divided into: group-1 (n = 45) with amount of plasmatic sEPCR below 100 ng/mL, group-2 (n = 45) where the concentration of sEPCR was between 100 and 200, and group-3 (n = 20) higher than 200 ng/mL. The numbers of thrombotic incidence recorded in each group were four, six, and eight, respectively. These results reveal that EPCR is expressed not only by a wide range of human malignant hematological cells but also the detection of plasma sEPCR levels provides a powerful insight into thrombotic risk assessment in cancer patients, especially when it surpasses 200 ng/mL.


Subject(s)
Antigens, CD/blood , Biomarkers, Tumor/genetics , Hematologic Neoplasms/blood , Receptors, Cell Surface/blood , Thrombophilia/blood , Antigens, CD/genetics , Base Sequence , Biomarkers, Tumor/blood , Cell Line, Tumor , Endothelial Protein C Receptor , Hematologic Neoplasms/complications , Humans , Polymorphism, Single Nucleotide , Receptors, Cell Surface/genetics , Retrospective Studies , Risk , Sequence Analysis, DNA , Thrombophilia/etiology
13.
Arterioscler Thromb Vasc Biol ; 32(2): 335-42, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22095979

ABSTRACT

OBJECTIVE: Calpains, calcium-activated proteases, mediate the angiogenic signals of vascular endothelial growth factor. However, their involvement in vascular repair has not been investigated and the underlying mechanisms remain to be fully elucidated. METHODS AND RESULTS: A rapidly progressive form of glomerulonephritis in wild type and transgenic mice expressing high levels of calpastatin, a calpain-specific inhibitor, was studied. Calpastatin transgene expression prevented the repair of peritubular capillaries and the recovery of renal function, limiting mouse survival. In vitro analysis detected a significant reduction of both intracellular and extracellular calpain activities in transgene expressing cells, whereas Western blotting revealed that proangiogenic factors vascular endothelial growth factor and norepinephrine increased calpain exteriorization. In vitro, extracellular calpains increased endothelial cell proliferation, migration and capillary tube formation. In vivo, delivery of nonpermeable extracellular calpastatin was sufficient to blunt angiogenesis and vascular repair. Endothelial cell response to extracellular calpains was associated with fibronectin cleavage, generating fibronectin fragments with proangiogenic capacity. In vivo, fibronectin cleavage was limited in the kidney of calpastatin transgenic mice with nephritis. CONCLUSIONS: This study demonstrates that externalized calpains participate in angiogenesis and vascular repair, partly by promoting fibronectin cleavage and thereby amplifying vascular endothelial growth factor efficiency. Thus, manipulation of calpain externalization may have therapeutic implications to control angiogenesis.


Subject(s)
Calpain/physiology , Disease Progression , Glomerulonephritis/physiopathology , Neovascularization, Physiologic/physiology , Animals , Blood Vessels/growth & development , Calpain/genetics , Calpain/pharmacology , Cell Movement/drug effects , Cell Movement/physiology , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Fibronectins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
14.
Cancer Lett ; 295(2): 242-51, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20359813

ABSTRACT

Matrix metalloproteinase-9 (MMP-9) strongly influences tumor development and metastasis. Using resistant (rMCF-7) and sensitive (sMCF-7) breast cancer lines we investigated the role of MMP-9 in cell migration (CM) and tubular network (TN) formation, two processes implied in tumor growth and metastasis. Our data demonstrate that MMP-9 which is critical for CM is necessary but not sufficient for TN formation and suggest a link between MDR1/P-gp and constitutive MMP-9. Both TN formation and CM are dependent on PKC and ERK1/2 pathways. This study reinforces the logic of combining therefore MMP inhibitors in cancer therapy, especially in patients with chemoresistance and invasion/metastasis.


Subject(s)
Breast Neoplasms/pathology , Matrix Metalloproteinase 9/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Protein Kinase C/physiology , Signal Transduction , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Drug Resistance, Neoplasm , Female , Flavonoids/pharmacology , Humans , Tetradecanoylphorbol Acetate/pharmacology
15.
Invest Ophthalmol Vis Sci ; 51(1): 421-9, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19661225

ABSTRACT

PURPOSE: Activated B-Raf alone cannot induce melanoma but must cooperate with other signaling pathways. The phosphatidylinositol 3-kinase (PI3K)/Akt and mammalian target of rapamycin (mTOR)/p70S6K pathways are critical for tumorigenesis. The authors investigated the role of these pathways in uveal melanoma cells. METHODS: The effects of PI3K and mTOR activation and inhibition on the proliferation of human uveal melanoma cell lines expressing either activated (WT)B-Raf or (V600E)B-Raf were investigated. Interactions among PI3K, mTOR, and B-Raf/ERK were studied. RESULTS: Inhibition of PI3K deactivated P70S6 kinase, reduced cell proliferation by 71% to 84%, and increased apoptosis by a factor of 5.0 to 8.4 without reducing ERK1/2 activation, indicating that ERK plays no role in mediating PI3K in these processes. In contrast, rapamycin-induced inhibition of mTOR did not significantly affect cell proliferation because it simultaneously stimulated PI3K/Akt activation and cyclin D1 expression. Regardless of B-Raf mutation status, cotreatment with the PI3K inhibitor effectively sensitized all melanoma cell lines to the B-Raf or ERK1/2 inhibition-induced reduction of cell proliferation. B-Raf/ERK and PI3K signaling, but not mTOR signaling, converged to control cyclin D1 expression. Moreover, p70S6K required the activation of ERK1/2. These data demonstrate that PI3K/Akt and mTOR/P70S6K interact with B-Raf/ERK. CONCLUSIONS: Activated PI3K/Akt attenuates the inhibitory effects of rapamycin on cell proliferation and thus serves as a negative feedback mechanism. This finding suggests that rapamycin is unlikely to inhibit uveal melanoma growth. In contrast, targeting PI3K while inhibiting B-Raf/ERK may be a promising approach to reduce the proliferation of uveal melanoma cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Melanoma/metabolism , Phosphatidylinositol 3-Kinases/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/physiology , Uveal Neoplasms/metabolism , Apoptosis , Blotting, Western , Cell Cycle/physiology , Cell Proliferation , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Feedback, Physiological , Humans , Melanoma/pathology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tumor Cells, Cultured , Uveal Neoplasms/pathology
16.
BMC Cancer ; 9: 199, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19549303

ABSTRACT

BACKGROUND: AVE9633 is a new immunoconjugate comprising a humanized monoclonal antibody, anti-CD33 antigen, linked through a disulfide bond to the maytansine derivative DM4, a cytotoxic agent and potent tubulin inhibitor. It is undergoing a phase I clinical trial. Chemoresistance to anti-mitotic agents has been shown to be related, in part, to overexpression of ABC proteins. The aim of the present study was to investigate the potential roles of P-gp, MRP1 and BCRP in cytotoxicity in AVE9633-induced acute myeloid leukaemia (AML). METHODS: This study used AML cell lines expressing different levels of P-gp, MRP1 or BCRP proteins and twenty-five samples from AML patients. Expression and functionality of the transporter protein were analyzed by flow cytometry. The cytotoxicity of the drug was evaluated by MTT and apoptosis assays. RESULTS: P-gp activity, but not MRP1 and BCRP, attenuated AVE9633 and DM4 cytotoxicity in myeloid cell lines. Zosuquidar, a potent specific P-gp inhibitor, restored the sensitivity of cells expressing P-gp to both AVE9633 and DM4. However, the data from AML patients show that 10/25 samples of AML cells (40%) were resistant to AVE9633 or DM4 (IC(50) > 500 nM), and this was not related to P-gp activity (p-Value: 0.7). Zosuquidar also failed to re-establish drug sensitivity. Furthermore, this resistance was not correlated with CD33 expression (p-Value: 0.6) in those cells. CONCLUSION: P-gp activity is not a crucial mechanism of chemoresistance to AVE9633. For patients whose resistance to conventional anthracycline AML regimens is related to ABC protein expression, a combination with AVE9633 could be beneficial. Other mechanisms such as microtubule alteration could play an important role in chemoresistance to AVE9633.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Immunoconjugates/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Maytansine/analogs & derivatives , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antibodies, Monoclonal/pharmacology , Antigens, CD/biosynthesis , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/biosynthesis , Antigens, Differentiation, Myelomonocytic/immunology , Dibenzocycloheptenes/pharmacology , Drug Resistance, Neoplasm , HL-60 Cells , Humans , Immunoconjugates/pharmacokinetics , K562 Cells , Maytansine/pharmacokinetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , Quinolines/pharmacology , Sialic Acid Binding Ig-like Lectin 3
17.
Aging Cell ; 8(2): 162-77, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19239420

ABSTRACT

Age-related macular degeneration (AMD) is characterized by the formation of drusen, extracellular deposits associated with atrophy of the retinal pigmented epithelium (RPE), disturbance of the transepithelial barrier and photoreceptor death. Amyloid-beta (Abeta) is present in drusen but its role during AMD remains unknown. This study investigated the in vitro and in vivo effects of the oligomeric form of Abeta(1-42) - OAbeta(1-42) - on RPE and found that it reduced mitochondrial redox potential and increased the production of reactive oxygen species, but did not induce apoptosis in RPE cell cultures. It also disorganized the actin cytoskeleton and halved occludin expression, markedly decreasing attachment capacity and abolishing the selectivity of RPE cell transepithelial permeability. Antioxidant pretreatment partially reversed the effects of OAbeta(1-42) on mitochondrial redox potential and transepithelial permeability. Subretinally injected OAbeta(1-42) induced pigmentation loss and RPE hypertrophy but not RPE cell apoptosis in C57BL/6 J mice. Rapid OAbeta(1-42)-induced disorganization of cytoskeletal actin filaments was accompanied by decreased RPE expression of the tight junction proteins occludin and zonula occludens-1 and of the visual cycle proteins cellular retinaldehyde-binding protein and RPE65. The number of photoreceptors decreased by half within a few days. Our study pinpoints the role of Abeta in RPE alterations and dysfunctions leading to retinal degeneration and suggests that targeting Abeta may help develop selective methods for treating diseases involving retinal degeneration, such as AMD.


Subject(s)
Amyloid beta-Peptides/toxicity , Macular Degeneration/physiopathology , Oxidative Stress/drug effects , Peptide Fragments/toxicity , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Aging/metabolism , Aging/pathology , Amyloid beta-Peptides/metabolism , Animals , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/pathology , Eye Proteins/drug effects , Eye Proteins/metabolism , Humans , Hypertrophy/chemically induced , Hypertrophy/metabolism , Hypertrophy/physiopathology , Macular Degeneration/chemically induced , Macular Degeneration/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Peptide Fragments/metabolism , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/pathology , cis-trans-Isomerases
18.
Invest Ophthalmol Vis Sci ; 50(3): 1047-57, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19029025

ABSTRACT

PURPOSE: Constitutive activation of ERK1/2 controls proliferation of uveal melanoma cells. Because an autocrine fibroblast growth factor (FGF) activation loop controls ERK1/2 activation in many cancers, this study was conducted to examine the role of the FGF/FGF receptor autocrine loop in the ERK1/2-dependent proliferation and survival of uveal melanoma cells. METHODS: Primary tumors and cell lines (OCM-1, MKT-BR, SP6.5, Mel270 and 92.1) were used to define the role of the FGF/FGFR system in human uveal melanoma. Cell proliferation was assessed by MTT-staining, and apoptosis was quantified by flow cytometry. Specific pharmacologic inhibitors of ERK1/2 and FGFR1, an anti-FGF2 neutralizing antibody and an antisense oligonucleotide directed against FGF2 were used to analyze signaling in the FGF/FGFR autocrine loop. RESULTS: FGF1, FGF2, and their FGFR1 receptor were strongly expressed in the primary uveal melanomas. All five uveal melanoma cell lines expressed and secreted FGF2. They also expressed FGFR1. Cell proliferation was strongly reduced by the antisense oligonucleotide-mediated depletion of endogenous FGF2, immunoneutralization of secreted FGF2, and pharmacologic inhibition of FGFR1. The FGF2/FGFR1-mediated signaling pathway was identified by showing that inhibition of either FGF2 or FGFR1 reduced ERK1/2 activation, cell proliferation, and survival. CONCLUSIONS: The FGF/FGFR/ERK signaling pathway may be a target for therapeutic strategies against uveal melanoma.


Subject(s)
Autocrine Communication/physiology , Cell Proliferation , Fibroblast Growth Factor 2/metabolism , Melanoma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Uveal Neoplasms/metabolism , Apoptosis , Blotting, Western , Cell Survival , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Flow Cytometry , Humans , Melanoma/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 , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins B-raf/physiology , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tetrazolium Salts/metabolism , Thiazoles/metabolism , Thionucleotides/pharmacology , Tumor Cells, Cultured , Uveal Neoplasms/pathology
19.
PLoS One ; 3(12): e3894, 2008.
Article in English | MEDLINE | ID: mdl-19079610

ABSTRACT

BACKGROUND: The microenvironment plays a major role in the onset and progression of metastasis. Epithelial ovarian cancer (EOC) tends to metastasize to the peritoneal cavity where interactions within the microenvironment might lead to chemoresistance. Mesothelial cells are important actors of the peritoneal homeostasis; we determined their role in the acquisition of chemoresistance of ovarian tumours. METHODOLOGY/PRINCIPAL FINDINGS: We isolated an original type of stromal cells, referred to as "Hospicells" from ascitis of patients with ovarian carcinosis using limiting dilution. We studied their ability to confer chemoresistance through heterocellular interactions. These stromal cells displayed a new phenotype with positive immunostaining for CD9, CD10, CD29, CD146, CD166 and Multi drug resistance protein. They preferentially interacted with epithelial ovarian cancer cells. This interaction induced chemoresistance to platin and taxans with the implication of multi-drug resistance proteins. This contact enabled EOC cells to capture patches of the Hospicells membrane through oncologic trogocytosis, therefore acquiring their functional P-gp proteins and thus developing chemoresistance. Presence of Hospicells on ovarian cancer tissue micro-array from patients with neo-adjuvant chemotherapy was also significantly associated to chemoresistance. CONCLUSIONS/SIGNIFICANCE: This is the first report of trogocytosis occurring between a cancer cell and an original type of stromal cell. This interaction induced autonomous acquisition of chemoresistance. The presence of stromal cells within patient's tumour might be predictive of chemoresistance. The specific interaction between cancer cells and stromal cells might be targeted during chemotherapy.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms/pathology , Stromal Cells/pathology , Aged , Biological Assay , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cell Separation , Epithelium/pathology , Epithelium/ultrastructure , Female , Humans , Intracellular Membranes/metabolism , Middle Aged , Multidrug Resistance-Associated Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/ultrastructure , Phenotype , Stromal Cells/ultrastructure
20.
Exp Hematol ; 36(12): 1625-33, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922614

ABSTRACT

OBJECTIVE: To investigate the proapoptotic capacities of four arylcoumarin analogues of combretastatins on leukemic cells from B-cell chronic lymphocytic leukemia (CLL), a malignancy characterized by apoptosis deficiency. MATERIALS AND METHODS: The effects of the four compounds on several nuclear, membrane, and mitochondrial events of apoptosis and on expression of proteins controlling the apoptosis were analyzed after treatment of cultured CLL patients' cells. RESULTS: Treatment with all four compounds resulted in a dose-dependent internucleosomal DNA fragmentation, in stimulation of phosphatidylserine externalization, disruption of the mitochondrial transmembrane potential and caspase-3 activation. DNA fragmentation was prevented in the presence of the pan-caspase inhibitor z-VAD-fmk. Two of the compounds downregulated the expression of Mcl-1, a protein thought to be crucial for the antiapoptotic state in CLL, while Bcl-2 expression was unaffected. No effects were observed on the expression of p27kip1 or the inducible nitric oxide synthase, two proteins, which are constitutively overexpressed by CLL cells and downregulated during the apoptosis induced by other plant-derived molecules (flavopiridol, polyphenols, or hyperforin). This suggests different mechanisms of action for the compounds studied here. Furthermore, normal B lymphocytes from healthy donors appeared less sensitive than CLL cells to the proapoptotic activity of the four compounds. CONCLUSION: The four arylcoumarin analogues were able to promote the apoptosis of CLL cells ex vivo through the caspase-dependent mitochondrial pathway. Therefore, these compounds may be of interest to develop new therapies of CLL based on apoptosis restoration.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Bibenzyls/pharmacology , DNA Fragmentation/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Aged , Amino Acid Chloromethyl Ketones/pharmacology , Caspase 3/metabolism , Caspase Inhibitors , Cyclin-Dependent Kinase Inhibitor p27 , Cysteine Proteinase Inhibitors/pharmacology , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...