Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Pharmaceutics ; 15(3)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36986686

ABSTRACT

Dendritic cells (DCs) are the most specialized and proficient antigen-presenting cells. They bridge innate and adaptive immunity and display a powerful capacity to prime antigen-specific T cells. The interaction of DCs with the receptor-binding domain of the spike (S) protein from the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a pivotal step to induce effective immunity against the S protein-based vaccination protocols, as well as the SARS-CoV-2 virus. Herein, we describe the cellular and molecular events triggered by virus-like particles (VLPs) containing the receptor-binding motif from the SARS-CoV-2 spike protein in human monocyte-derived dendritic cells, or, as controls, in the presence of the Toll-like receptors (TLR)3 and TLR7/8 agonists, comprehending the events of dendritic cell maturation and their crosstalk with T cells. The results demonstrated that VLPs boosted the expression of major histocompatibility complex molecules and co-stimulatory receptors of DCs, indicating their maturation. Furthermore, DCs' interaction with VLPs promoted the activation of the NF-kB pathway, a very important intracellular signalling pathway responsible for triggering the expression and secretion of proinflammatory cytokines. Additionally, co-culture of DCs with T cells triggered CD4+ (mainly CD4+Tbet+) and CD8+ T cell proliferation. Our results suggested that VLPs increase cellular immunity, involving DC maturation and T cell polarization towards a type 1 T cells profile. By providing deeper insight into the mechanisms of activation and regulation of the immune system by DCs, these findings will enable the design of effective vaccines against SARS-CoV-2.

2.
Cell Mol Life Sci ; 79(4): 213, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35344105

ABSTRACT

Endoplasmic reticulum (ER) stress and mitochondrial dysfunction, which are key events in the initiation and/or progression of several diseases, are correlated with alterations at ER-mitochondria contact sites, the so-called "Mitochondria-Associated Membranes" (MAMs). These intracellular structures are also implicated in NLRP3 inflammasome activation which is an important driver of sterile inflammation, however, the underlying molecular basis remains unclear. This work aimed to investigate the role of ER-mitochondria communication during ER stress-induced NLRP3 inflammasome activation in both peripheral and central innate immune systems, by using THP-1 human monocytes and BV2 microglia cells, respectively, as in vitro models. Markers of ER stress, mitochondrial dynamics and mass, as well as NLRP3 inflammasome activation were evaluated by Western Blot, IL-1ß secretion was measured by ELISA, and ER-mitochondria contacts were quantified by transmission electron microscopy. Mitochondrial Ca2+ uptake and polarization were analyzed with fluorescent probes, and measurement of aconitase and SOD2 activities monitored mitochondrial ROS accumulation. ER stress was demonstrated to activate the NLRP3 inflammasome in both peripheral and central immune cells. Studies in monocytes indicate that ER stress-induced NLRP3 inflammasome activation occurs by a Ca2+-dependent and ROS-independent mechanism, which is coupled with upregulation of MAMs-resident chaperones, closer ER-mitochondria contacts, as well as mitochondrial depolarization and impaired dynamics. Moreover, enhanced ER stress-induced NLRP3 inflammasome activation in the immune system was found associated with pathological conditions since it was observed in monocytes derived from bipolar disorder (BD) patients, supporting a pro-inflammatory status in BD. In conclusion, by demonstrating that ER-mitochondria communication plays a key role in the response of the innate immune cells to ER stress, this work contributes to elucidate the molecular mechanisms underlying NLRP3 inflammasome activation under stress conditions, and to disclose novel potential therapeutic targets for diseases associated with sterile inflammation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Endoplasmic Reticulum Stress , Humans , Immune System , Mitochondria
3.
Sci Rep ; 9(1): 18972, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831825

ABSTRACT

The aim is to study risk factors for retinal vein occlusion (RVO), such as thrombophilic and cardiovascular risk factors (CRF). A retrospective consecutive case series of 60 patients with RVO was made, tested for CRF, hyperhomocysteinemia, lupic anticoagulant, antiphospholipid antibody and 5 gene variants: factor V (FV) Leiden (G1691A), factor II (PT G20210A), 5,1-methylenetetra-hydrofolate reductase (MTHFR; 677 C > T and 1298 A > C), plasminogen activator inhibitor 1 (PAI-1; 4 G/5 G). More than 1 CRF were present in 36 patients (60%), which had a significantly higher mean age at diagnosis (66.7 ± 12.9 versus 59.5 ± 13.7 with ≤1 CRF, [t(57) = -2.05, p = 0.045, d = 0.54). Patients with thermolabile MTHFR forms with decreased enzyme activity (T677T or C677T/A1298C) had a significant lower mean age [57.6 ± 15.1; t (58) = 3.32; p = 0.002; d = 0.846] than patients with normal MTHFR enzyme activity (68.5 ± 10.2). Regarding CRF and thermolabile forms of MTHFR, the mean age at diagnosis could be significantly predicted [F(2,56) = 7.18; p = 0.002] by the equation: 64.8 - 10.3 × (thermolabile MTHFR) - 5.31 × ( ≤ 1CRF). Screening of MTHFR polymorphisms may be useful in younger RVO patients, particularly when multiple CRF are absent.


Subject(s)
Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Genetic , Retinal Vein Occlusion , Thrombophilia , Adult , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Retinal Vein Occlusion/enzymology , Retinal Vein Occlusion/etiology , Retinal Vein Occlusion/genetics , Risk Factors , Thrombophilia/complications , Thrombophilia/enzymology , Thrombophilia/genetics
6.
Eye Vis (Lond) ; 5: 27, 2018.
Article in English | MEDLINE | ID: mdl-30386806

ABSTRACT

OBJECTIVE: To evaluate short-term markers of outcome in diabetic macular edema (DME). METHODS: Prospective interventional case series included 122 eyes of 122 patients with recently diagnosed DME. Eyes were treated with a 3-monthly loading dose of ranibizumab or aflibercept and pro re nata thereafter. Serial enhanced deep imaging SD-OCT high resolution scans were used to measure subfoveal choroidal thickness (SFCT) and central retinal thickness (CRT). Anatomic (10% CRT decrease) and functional responses (best corrected visual acuity, BCVA gain ≥5 letters) were assessed at 3 months and 6 months using univariate and multivariate analyses. Parameters tested were gender, duration of diabetes, HbA1c, hypertension, CRT, SFCT, BCVA, ellipsoid zone (EZ) status, subfoveal neuroretinal detachment (SND), anti-VEGF used and laser naivety. A logistic regression model was applied to find independent markers outcome. RESULTS: BCVA increased, CRT and SFCT decreased at 3 months and 6 months. Good metabolic control (p = 0.003), intact baseline EZ (p = 0.030), EZ re-grading at 3 M (p < 0.001) and laser naivety (p = 0.001) were associated with better functional outcome. The multivariate linear regression model showed that baseline SND and CRT are predictors of anatomic response, while lower baseline BCVA and intact EZ are predictors of functional response. CONCLUSION: The presence of SND predicts anatomic response only, while an intact EZ is critical to achieve a good functional outcome in DME.

7.
Graefes Arch Clin Exp Ophthalmol ; 256(10): 1857-1865, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30039271

ABSTRACT

PURPOSE: The aim of this study was to evaluate subfoveal choroidal thickness (SFCT) as a marker of outcome in real-world treatment of diabetic macular edema (DME) and to correlate it with choroidal thicknesses (CT) collected around the fovea. METHODS: Prospective interventional case series included a total of 126 eyes from 126 patients with recently diagnosed DME treated with a 3-monthly loading dose of ranibizumab or aflibercept and PRN thereafter until 24 months (M). CT was manually measured in the central 3500 µm area, subfoveally (SFCT), at 1750 µm right and left from the center in the horizontal plane and at 1750 µm up and down from the center in the vertical plane, by OCT. Anatomic (10% decrease in central retinal thickness) and functional (gain ≥ 5 letters) responses were assessed using univariate and multivariate analyses. The areas under ROC curves were used to assess whether baseline SFCT was a predictor of outcome. RESULTS: CT significantly decreased in all follow-ups (3 months after the 3 injections' loading dose (3M), 6 months (6M), 12 months (12M), 18 months (18M), 24 months (24M)). SFCT and other CT parameters are correlated. SFCT decrease from baseline was related with treatment (p = 0.003 to p < 0.001) but not with anatomic (3M, p = 0.858; 6M p = 0.762) or functional response (3M, p = 0.746; 6M, p = 0.156). SFCT was not found to be predictive of anatomic (AUC = 0.575, p = 0.172) or functional (AUC = 0.515, p = 0.779) outcome. CONCLUSIONS: SFCT is a reliable marker of choroidal thickness. Baseline SFCT decreased with anti-VEGF treatment but did not predict DME outcome.


Subject(s)
Bevacizumab/administration & dosage , Choroid/pathology , Diabetic Retinopathy/drug therapy , Fluorescein Angiography/methods , Macular Edema/drug therapy , Receptors, Vascular Endothelial Growth Factor/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Tomography, Optical Coherence/methods , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Diabetic Retinopathy/complications , Diabetic Retinopathy/diagnosis , Female , Fovea Centralis/pathology , Fundus Oculi , Humans , Intravitreal Injections , Macular Edema/diagnosis , Macular Edema/etiology , Male , Middle Aged , Prospective Studies , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Visual Acuity
8.
Exp Cell Res ; 370(1): 68-77, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29902537

ABSTRACT

One of the major challenges in Glioblastoma (GBM) therapy relates with the existence of glioma stem-like cells (GSCs), known to be chemo- and radio-resistant. GSCs and non-stem GBM cells have the ability to interchange, emphasizing the importance of identifying common molecular targets among those cell sub-populations. Nucleolin overexpression has been recently associated with breast cancer sub-populations with different stem-like phenotype. The goal of this work was to evaluate the potential of cell surface nucleolin as a target in GBM cells. Different levels of nucleolin expression resulted in a 3.4-fold higher association of liposomes targeting nucleolin (functionalized with the nucleolin-binding F3 peptide) in U87, relative to GBM11 glioblastoma cells. Moreover, nucleolin was suggested as a potential marker in OCT4-, NANOG-positive GSC, and in the corresponding non-stem GBM cells, as well as in SOX2-positive GSC. Doxorubicin delivered by liposomes targeting nucleolin enabled a level of cytotoxicity that was 2.5- or 4.6-fold higher compared to the non-targeted counterparts. Importantly, an overexpression of nucleolin was also observed in cells of patient-derived samples, as compared with normal brain. Overall, these results suggested nucleolin as a therapeutic target in GBM.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cytotoxins/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Biomarkers, Tumor/metabolism , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Glioma/drug therapy , Glioma/metabolism , Humans , Liposomes/pharmacology , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Nucleolin
9.
Oncol Rep ; 38(3): 1341-1352, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714013

ABSTRACT

Glioblastoma (GBM) is a grade IV astrocytoma. GBM patients show resistance to chemotherapy such as temozolomide (TMZ), the gold standard treatment. In order to simulate the molecular mechanisms behind the different chemotherapeutic responses in GBM patients we compared the cellular heterogeneity and chemotherapeutic resistance mechanisms in different GBM cell lines. We isolated and characterized a human GBM cell line obtained from a GBM patient, named GBM11. We studied the GBM11 behaviour when treated with Tamoxifen (TMX) that, among other functions, is a protein kinase C (PKC) inhibitor, alone and in combination with TMZ in comparison with the responses of U87 and U118 human GBM cell lines. We evaluated the cell death, cell cycle arrest and cell proliferation, mainly through PKC expression, by flow cytometry and western blot analysis and, ultimately, cell migration capability and f-actin filament disorganization by fluorescence microscopy. We demonstrated that the constitutive activation of p-PKC seems to be one of the main metabolic implicated on GBM malignancy. Despite of its higher resistance, possibly due to the overexpression of P-glycoprotein and stem-like cell markers, GBM11 cells presented a subtle different chemotherapeutic response compared to U87 and U118 cells. The GBM11, U87, U118 cell lines show subtle molecular differences, which clearly indicate the characterization of GBM heterogeneity, one of the main reasons for tumor resistance. The adding of cellular heterogeneity in molecular behaviour constitutes a step closer in the understanding of resistant molecular mechanisms in GBM, and can circumvents the eventual impaired therapy.


Subject(s)
Astrocytoma/drug therapy , Genetic Heterogeneity , Glioblastoma/drug therapy , Protein Kinase C/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Apoptosis/drug effects , Astrocytoma/genetics , Astrocytoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Neoplasm Grading , Signal Transduction/drug effects , Tamoxifen/administration & dosage , Temozolomide
10.
Transl Oncol ; 10(4): 555-569, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28654819

ABSTRACT

Glioblastoma (GBM) is the most malignant primary brain tumor, with an average survival rate of 15 months. GBM is highly refractory to therapy, and such unresponsiveness is due, primarily, but not exclusively, to the glioma stem-like cells (GSCs). This subpopulation express stem-like cell markers and is responsible for the heterogeneity of GBM, generating multiple differentiated cell phenotypes. However, how GBMs maintain the balance between stem and non-stem populations is still poorly understood. We investigated the GBM ability to interconvert between stem and non-stem states through the evaluation of the expression of specific stem cell markers as well as cell communication proteins. We evaluated the molecular and phenotypic characteristics of GSCs derived from differentiated GBM cell lines by comparing their stem-like cell properties and expression of connexins. We showed that non-GSCs as well as GSCs can undergo successive cycles of gain and loss of stem properties, demonstrating a bidirectional cellular plasticity model that is accompanied by changes on connexins expression. Our findings indicate that the interconversion between non-GSCs and GSCs can be modulated by extracellular factors culminating on differential expression of stem-like cell markers and cell-cell communication proteins. Ultimately, we observed that stem markers are mostly expressed on GBMs rather than on low-grade astrocytomas, suggesting that the presence of GSCs is a feature of high-grade gliomas. Together, our data demonstrate the utmost importance of the understanding of stem cell plasticity properties in a way to a step closer to new strategic approaches to potentially eliminate GSCs and, hopefully, prevent tumor recurrence.

11.
Cell Oncol (Dordr) ; 40(3): 247-261, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28401486

ABSTRACT

PURPOSE: Glioblastomas (GBM) comprise 17% of all primary brain tumors. These tumors are extremely aggressive due to their infiltrative capacity and chemoresistance, with glial-to-mesenchymal transition (GMT) proteins playing a prominent role in tumor invasion. One compound that has recently been used to reduce the expression of these proteins is shikonin (SHK), a naphthoquinone with anti-tumor properties. Temozolomide (TMZ), the most commonly used chemotherapeutic agent in GBM treatment, has so far not been studied in combination with SHK. Here, we investigated the combined effects of these two drugs on the proliferation and motility of GBM-derived cells. METHODS: The cytotoxic and proliferative effects of SHK and TMZ on human GBM-derived cells were tested using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), Ki67 staining and BrdU incorporation assays. The migration capacities of these cells were evaluated using a scratch wound assay. The expression levels of ß3 integrin, metalloproteinases (MMPs) and GMT-associated proteins were determined by Western blotting and immunocytochemistry. RESULTS: We found that GBM-derived cells treated with a combination of SHK and TMZ showed decreases in their proliferation and migration capacities. These decreases were followed by the suppression of GMT through a reduction of ß3 integrin, MMP-2, MMP-9, Slug and vimentin expression via inactivation of PI3K/AKT signaling. CONCLUSION: From our results we conclude that dual treatment with SHK and TMZ may constitute a powerful new tool for GBM treatment by reducing therapy resistance and tumor recurrence.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioblastoma/pathology , Naphthoquinones/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Temozolomide
12.
Mycopathologia ; 181(11-12): 879-884, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27631818

ABSTRACT

We report a case of a keratitis associated with a Fusarium penzigii-a Fusarium dimerum species complex (FDSC)-in a 81-year-old woman after a corneal trauma with a tree branch. At patient admittance, slit lamp biomicroscopy revealed an exuberant chemosis, an inferior corneal ulcer with an associated inflammatory infiltrate, a central corneal abscess, bullous keratopathy and posterior synechiae. Corneal scrapes were obtained for identification of bacteria and fungi, and the patient started antibiotic treatment on empirical basis. Few days later, the situation worsened with the development of hypopyon. By that time, Fusarium was identified in cultures obtained from corneal scrapes and the patient started topical amphotericin B 0.15 %. Upon the morphological identification of the Fusarium as a FDSC, and since there was no clinical improvement, the treatment with amphotericin B was suspended and the patient started voriconazole 10 mg/ml, eye drops, hourly and voriconazole 200 mg iv, every 12 h for 1 month. The hypopyon resolved and the inflammatory infiltrate improved, but the abscess persisted at the last follow-up visit. The molecular identification revealed that the FDSC was a F. penzigii.


Subject(s)
Corneal Injuries/complications , Fusariosis/diagnosis , Fusariosis/pathology , Fusarium/isolation & purification , Keratitis/etiology , Keratitis/pathology , Aged, 80 and over , Antifungal Agents/administration & dosage , Female , Fusariosis/microbiology , Fusarium/classification , Humans , Keratitis/microbiology , Ophthalmic Solutions/administration & dosage , Treatment Outcome , Voriconazole/administration & dosage
13.
Biochim Biophys Acta ; 1850(4): 722-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25554223

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a highly proliferative, angiogenic grade IV astrocytoma that develops resistance to the alkylating agents used in chemotherapy, such as temozolomide (TMZ), which is considered the gold standard. The mean survival time for GBM patients is approximately 12 months, increasing to 14.6 months after TMZ treatment. The resistance of GBM to chemotherapy seems to be associated to genetic alterations and to the constitutive activation of several signaling pathways. Therefore, the combination of different drugs with different mechanisms of action may contribute to circumvent the chemoresistance of glioma cells. Here we describe the potential synergistic behavior of the therapeutic combination of tamoxifen (TMX), a known inhibitor of PKC, and TMZ in GBM. METHODS: We used two GBM cell lines incubated in absence and presence of TMX and/or TMZ and measured cell viability, proliferation, apoptosis, cell cycle, migration ability, cytoskeletal organization and the phosphorylated amount of the p-PKC-pan. RESULTS: The combination of low doses of TMX with increasing doses of TMZ shows an increased antiproliferative and apoptotic effect compared to the effect with TMX alone. CONCLUSIONS: The combination of TMX and TMZ seems to potentiate the effect of each other. These alterations seem to be associated to a decrease in the phosphorylation status of PKC. GENERAL SIGNIFICANCE: We emphasize that TMX is an inhibitor of the p-PKC-pan and that these combination is more effective in the reduction of proliferation and in the increase of apoptosis than each drug alone, which presents a new therapeutic strategy in GBM treatment.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/enzymology , Protein Kinase C/antagonists & inhibitors , Tamoxifen/pharmacology , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Drug Synergism , Glioblastoma/pathology , Humans , Phosphorylation , Temozolomide
14.
Cancer Biol Ther ; 14(4): 287-94, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23358475

ABSTRACT

Glioblastoma Multiforme (GBM) is the most aggressive brain tumor characterized by intratumoral heterogeneity at cytopathological, genomic and transcriptional levels. Despite the efforts to develop new therapeutic strategies the median survival of GBM patients is 12-14 months. Results from large-scale gene expression profile studies confirmed that the genetic alterations in GBM affect pathways controlling cell cycle progression, cellular proliferation and survival and invasion ability, which may explain the difficulty to treat GBM patients. One of the signaling pathways that contribute to the aggressive behavior of glioma cells is the protein kinase C (PKC) pathway. PKC is a family of serine/threonine-specific protein kinases organized into three groups according the activating domains. Due to the variability of actions controlled by PKC isoforms, its contribution to the development of GBM is poorly understood. This review intends to highlight the contribution of PKC isoforms to proliferation, survival and invasive ability of glioma cells.


Subject(s)
Brain Neoplasms/enzymology , Glioblastoma/enzymology , Protein Kinase C/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Protein Kinase C/genetics , Signal Transduction
15.
BMC Cancer ; 12: 139, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22475227

ABSTRACT

BACKGROUND: Osteosarcoma is a bone-forming tumor of mesenchymal origin that presents a clinical pattern that is consistent with the cancer stem cell model. Cells with stem-like properties (CSCs) have been identified in several tumors and hypothesized as the responsible for the relative resistance to therapy and tumor relapses. In this study, we aimed to identify and characterize CSCs populations in a human osteosarcoma cell line and to explore their role in the responsiveness to conventional therapies. METHODS: CSCs were isolated from the human MNNG/HOS cell line using the sphere formation assay and characterized in terms of self-renewal, mesenchymal stem cell properties, expression of pluripotency markers and ABC transporters, metabolic activity and tumorigenicity. Cell's sensitivity to conventional chemotherapeutic agents and to irradiation was analyzed and related with cell cycle-induced alterations and apoptosis. RESULTS: The isolated CSCs were found to possess self-renewal and multipotential differentiation capabilities, express markers of pluripotent embryonic stem cells Oct4 and Nanog and the ABC transporters P-glycoprotein and BCRP, exhibit low metabolic activity and induce tumors in athymic mice. Compared with parental MNNG/HOS cells, CSCs were relatively more resistant to both chemotherapy and irradiation. None of the treatments have induced significant cell-cycle alterations and apoptosis in CSCs. CONCLUSIONS: MNNG/HOS osteosarcoma cells contain a stem-like cell population relatively resistant to conventional chemotherapeutic agents and irradiation. This resistant phenotype appears to be related with some stem features, namely the high expression of the drug efflux transporters P-glycoprotein and BCRP and their quiescent nature, which may provide a biological basis for resistance to therapy and recurrence commonly observed in osteosarcoma.


Subject(s)
Bone Neoplasms/therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , Osteosarcoma/therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Biomarkers, Tumor/metabolism , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Flow Cytometry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Reactive Oxygen Species/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Spheroids, Cellular/radiation effects , Tumor Cells, Cultured
16.
Rheumatology (Oxford) ; 50(9): 1569-77, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21586522

ABSTRACT

OBJECTIVES: CD38 participates in lymphocyte ontogeny and function and may be involved in autoimmunity. Absence of CD38 accelerates development of non-obese diabetic (NOD) mice diabetes and anti-CD38 antibodies are good markers of human disease. Little is known regarding systemic autoimmunity. Active SLE patients have higher numbers of CD38(+) T and B cells. CD38 is a candidate gene for the murine Lmb2 lupus locus. We aimed to investigate whether CD38 was involved in lupus development. METHODS: We developed Cd38(-/-)-Fas(lpr)/Fas(lpr) mice and monitored them for development of a lupus-like disease through measurement of protein excretion in urine, histological assessment of the kidneys, quantification of circulating immunoglobulins and autoantibodies. We have also immunophenotyped 2- and 6-month old Cd38(-/-)-Fas(lpr)/Fas(lpr) mice. RESULTS: We found that absence of CD38 accelerated disease development: female Cd38(-/-)-Fas(lpr)/Fas(lpr) mice presented severe proteinuria, GN, deposition of ICs in the renal medulla and increased amounts of circulating immunoglobulin G (IgG), although anti-dsDNA autoantibodies and RF were not significantly increased at disease onset. We have found that Cd38(-/-)-Fas(lpr)/Fas(lpr) male mice, similarly to other murine models of lupus, were able to control disease. Absence of CD38 in lpr mice altered differentiation of T cells and dendritic cells (DC). CONCLUSION: Although the role of CD38 in tolerance is still to be elucidated, we provide evidence that it may play an active role in the control of a murine lupus-like disease.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Autoantibodies/immunology , Disease Models, Animal , Immunoglobulin G/immunology , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/immunology , Animals , Autoantibodies/metabolism , Female , Immunoglobulin G/metabolism , Kidney/pathology , Lymph Nodes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Proteinuria/etiology , Sex Factors , Spleen/immunology , Thymus Gland/immunology
18.
Cancer Biol Ther ; 9(1): 56-65, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19923906

ABSTRACT

Glioblastoma (GBM) is the most aggressive and malignant brain tumor. Recent studies indicated that glioma samples are characterized by increased expression of CXCR4, the CXCL12/SDF-1 chemokine receptor. To better understand the role of CXCR4 in GBM biology we performed an integrated study where we simultaneously evaluate the contribution of the CXCR4/CXCL12 signaling pathway to the proliferation, survival and motility of a human GBM cell line. Our results indicated that CXCR4/CXCL12 axis induced an increase in cell proliferation and in cell motility. The blockage of CXCR4 induced a significant increase of apoptosis. Together, our results indicated that CXCR4/CXCL12 signalling pathway may contribute to GBM development and emphasize the therapeutic potential of this pathway in patients with GBM.


Subject(s)
Brain Neoplasms/genetics , Cell Proliferation , Chemokine CXCL12/metabolism , Glioma/genetics , Receptors, CXCR4/metabolism , Cell Line, Tumor , Cell Movement/genetics , Chemokine CXCL12/genetics , Chemotaxis , Glioblastoma/pathology , Humans , Receptors, CXCR4/genetics
19.
Microbes Infect ; 9(7): 847-54, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17533152

ABSTRACT

CD38 is a multifunctional ectoenzyme that behaves either as an enzyme, a cell adhesion molecule or as a cell surface receptor involved in cell signalling. It is expressed in cells of several lineages, including B and T lymphocytes, and macrophages. CD38 was shown to be important for the development of T-cell dependent humoral immune responses against extracellular pathogens. It also appears to be functionally important in macrophages, which are the host cells of Mycobacterium avium, an intracellular parasite that survives within these cells by avoiding a number of their microbicidal strategies. The present work aimed at investigating whether CD38 had any role on the immune response against mycobacterial infection. After intraperitoneal M. avium infection, the immune response of CD38KO mice was compared to that of their parental strain, C57Bl.6 mice. Absence of CD38 rendered mice more susceptible to mycobacterial infection. This susceptibility seems to be due to ineffective Th1 differentiation and polarization, which is essential for the control of M. avium infection. In addition, absence of CD38 seems to compromise the maintenance of the granulomatous barrier, leading to dissemination and unrestrained growth of mycobacteria.


Subject(s)
ADP-ribosyl Cyclase 1/immunology , Granuloma/microbiology , Mycobacterium avium/immunology , Th1 Cells/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Animals , Enzyme-Linked Immunosorbent Assay , Female , Granuloma/immunology , Histocytochemistry , Interferon-gamma/immunology , Interleukin-4/immunology , Liver/immunology , Liver/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/microbiology , Th1 Cells/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...