Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 20(9)2019 Apr 30.
Article in English | MEDLINE | ID: mdl-31052281

ABSTRACT

Glioblastoma (GBM) is the most malignant tumor type affecting the adult central nervous system. Despite advances in therapy, the prognosis for patients with GBM remains poor, with a median survival of about 15 months. To date, few treatment options are available and recent trials based on the molecular targeting of some of the GBM hallmark pathways (e.g., angiogenesis) have not produced any significant improvement in overall survival. The urgent need to develop more efficacious targeted therapies has led to a better molecular characterization of GBM, revealing an emerging role of semaphorins in GBM progression. Semphorins are a wide group of membrane-bound and secreted proteins, originally identified as axon guidance cues, signaling through their receptors, neuropilins, and plexins. A number of semaphorin signals involved in the control of axonal growth and navigation during development have been found to furthermore participate in crosstalk with different dysfunctional GBM pathways, controlling tumor cell proliferation, migration, and invasion, as well as tumor angiogenesis or immune response. In this review, we summarize the regulatory activities mediated by semaphorins and their receptors on the oncogenic pathways implicated in GBM growth and invasive/metastatic progression.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Semaphorins/metabolism , Animals , Brain Neoplasms/pathology , Glioblastoma/pathology , Humans , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Semaphorins/genetics
2.
Cancer Cell Int ; 18: 136, 2018.
Article in English | MEDLINE | ID: mdl-30214378

ABSTRACT

BACKGROUND: Glioblastoma multiforme (GBM) is a highly aggressive brain tumor in which cancer cells with stem cell-like features, called cancer stem cells (CSCs), were identified. Two CSC populations have been previously identified in GBM, one derived from the GBM area called enhanced lesion (GCSCs) and the other one from the brain area adjacent to the tumor margin (PCSCs) that greatly differ in their growth properties and tumor-initiating ability. To date the most effective chemotherapy to treat GBM is represented by alkylating agents such as temozolomide (TMZ), whose activity can be regulated by histone deacetylases (HDACs) inhibitors through the modulation of O6-methylguanine-DNA methyltransferase (MGMT) expression. Levetiracetam (LEV), a relatively new antiepileptic drug, modulates HDAC levels ultimately silencing MGMT, thus increasing TMZ effectiveness. However, an improvement in the therapeutic efficacy of TMZ is needed. METHODS: Cell proliferation was investigated by BrdU cell proliferation assay and by Western Blot analysis of PCNA expression. Apoptosis was evaluated by Western Blot and Immunofluorescence analysis of the cleaved Caspase-3 expression. MGMT and HDAC4 expression was analyzed by Western Blotting and Immunofluorescence. Statistical analysis was performed using the Student's t test and Mann-Whitney test. RESULTS: Here we evaluated the effect of TMZ on the proliferation rate of the IDH-wildtype GCSCs and PCSCs derived from six patients, in comparison with the effects of other drugs such as etoposide, irinotecan and carboplatin. Our results demonstrated that TMZ was less effective compared to the other agents; hence, we verified the possibility to increase the effect of TMZ by combining it with LEV. Here we show that LEV enhances the effect of TMZ on GCSCs proliferation (being less effective on PCSCs) by decreasing MGMT expression, promoting HDAC4 nuclear translocation and activating apoptotic pathway. CONCLUSIONS: Although further studies are needed to determine the exact mechanism by which LEV makes GBM stem cells more  sensitive to TMZ, these results suggest that the clinical therapeutic efficacy of TMZ in GBM might be enhanced by the combined treatment with LEV.

3.
Oncotarget ; 9(46): 28116-28130, 2018 Jun 15.
Article in English | MEDLINE | ID: mdl-29963265

ABSTRACT

In glioblastoma multiforme (GBM), cancer stem cells (CSCs) are thought to be responsible for gliomagenesis, resistance to treatment and recurrence. Unfortunately, the prognosis for GBM remains poor and recurrence frequently occurs in the peritumoral tissue within 2 cm from the tumor edge. In this area, a population of CSCs has been demonstrated which may recapitulate the tumor after surgical resection. In the present study, we aimed to characterize CSCs derived from both peritumoral tissue (PCSCs) and GBM (GCSCs) in order to deepen their significance in GBM development and progression. The stemness of PCSC/GCSC pairs obtained from four human GBM surgical specimens was investigated by comparing the expression of specific stem cell markers such as Nestin, Musashi-1 and SOX2. In addition, the growth rate, the ultrastructural features and the expression of other molecules such as c-Met, pMet and MAP kinases, involved in cell migration/invasion, maintenance of tumor stemness and/or resistance to treatments were evaluated. Since it has been recently demonstrated the involvement of the long non-coding RNAs (lncRNAs) in the progression of gliomas, the expression of H19 lncRNA, as well as of one of its two mature products miR-675-5p was evaluated in neurospheres. Our results show significant differences between GCSCs and PCSCs in terms of proliferation, ultrastructural peculiarities and, at a lower extent, stemness profile. These differences might be important in view of their potential role as a therapeutic target.

4.
Arthritis Res Ther ; 20(1): 75, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29669578

ABSTRACT

BACKGROUND: The purpose of this study was to investigate the frequency and the distribution of inflammatory cell infiltrate in two sets of cutaneous biopsies derived from clinically affected and unaffected skin in patients with systemic sclerosis (SSc) and to test correlation between the cell infiltrate and the progression of skin involvement. METHODS: Skin was immunohistochemically assessed to identify CD68, CD3, CD20 and CD138-positive (+) cells in clinically affected and unaffected skin in 28 patients with SSc. Patients were followed for 6 months and the characteristics of the infiltrate were analyzed according to disease duration, clinical features and skin involvement progression. RESULTS: In all SSc cutaneous specimens, cellular infiltrates were found in a perivascular location predominantly in the mid and deeper portions of the dermis. All the analyzed biopsies showed a CD3+ and CD68+ cell infiltrate and the mean number of CD3+ and of CD68+ cells was higher in clinically involved skin (CD3+, 71.7 ± 34.6 and CD68+, 26.3 ± 8.4, respectively) than in clinically uninvolved skin (CD3+, 45.7 ± 36.0 and CD68+, 13.6 ± 6.1, respectively) (p < 0.001 for both comparisons). CD20+ cells were found in 17 (60.7%) patients and in these patients the mean number of CD20+ cells was higher in clinically involved (4.7 ± 5.9) than in uninvolved skin (1.9 ± 2.9), (p = 0.04). There was a greater number of CD20+ cells in patients with early SSc compared with patients with long-standing disease. CD138+ cells were found in 100% of biopsies of clinically involved skin and in 89.3% of biopsies of uninvolved skin. The mean number of CD138+ cells was higher in clinically involved skin (3.6 ± 2.3) than in clinically uninvolved skin (1.9 ± 1.7), (p < 0.001). Seven patients experienced more than 20% worsening in the skin score after 6 months of follow up; all of them had a CD20+ skin infiltrate on biopsy of clinically involved skin. CONCLUSIONS: Our results confirm that mononuclear cells are present in the skin of all patients with SSc, underlining the role of inflammatory cell infiltrates in skin involvement in SSc. B cells in the skin seem to characterize patients with early diffuse skin disease and to correlate with skin progression.


Subject(s)
B-Lymphocytes , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Skin/immunology , Skin/pathology , Adult , Aged , B-Lymphocytes/immunology , Disease Progression , Female , Humans , Macrophages/immunology , Male , Middle Aged , T-Lymphocytes/immunology , Young Adult
5.
Oncotarget ; 7(48): 78541-78556, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27705944

ABSTRACT

The formation of new blood vessels represents a crucial event under both physiological and pathological circumstances. In this study, we evaluated by immunohistochemistry, and/or Western blotting and/or quantitative real time-PCR the expression of HIF1α, HIF2α, VEGF, VEGFR1 and VEGFR2 in surgical glioblastoma multiforme (GBM) and peritumoral tissue samples obtained from 50 patients as well as in cancer stem cells (CSCs) isolated from GBM (GCSCs) and peritumoral tissue (PCSCs) of 5 patients. We also investigated the contribution of both GCSCs and PCSCs on the behavior of endothelial cells (ECs) in vitro. Immunohistochemistry demonstrated the expression of angiogenesis markers in both GBM and peritumoral tissue. In addition, in vitro tube formation assay indicated that both GCSCs and PCSCs stimulate EC proliferation as well as tube-like vessel formation. An increased migration aptitude was mainly observed when ECs were cultured in the presence of GCSCs rather than in the presence of PCSCs. These findings suggest that relevant neoangiogenetic events may occur in GBM. In particular, VEGF/VEGFR co-expression in PCSCs leads to hypothesize the involvement of an autocrine signaling. Moreover, our results suggest that both GCSCs and PCSCs own the skill of activating the "angiogenic switch" and the capability of modulating EC behavior, indicating that both cell types are either responsive to angiogenic stimuli or able to trigger angiogenic response. Together with our previous findings, this study adds a further piece to the challenging puzzle of the characterization of peritumoral tissue and of the definition of its real role in GBM pathophysiology.


Subject(s)
Angiogenic Proteins/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic , Adult , Aged , Angiogenic Proteins/genetics , Autocrine Communication , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Movement , Cell Proliferation , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/therapy , Human Umbilical Vein Endothelial Cells/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Neoplastic Stem Cells/pathology , Signal Transduction , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Young Adult
6.
J Neuropathol Exp Neurol ; 75(2): 134-47, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26792897

ABSTRACT

Characterization of tissue surrounding glioblastoma (GBM) is a focus for translational research because tumor recurrence invariably occurs in this area. We investigated the expression of the progenitor/stem cell markers GD3 ganglioside and NG2 proteoglycan in GBM, peritumor tissue (brain adjacent to tumor, BAT) and cancer stem-like cells (CSCs) isolated from GBM (GCSCs) and BAT (PCSCs). GD3 and NG2 immunohistochemistry was performed in paired GBM and BAT specimens from 40 patients. Double-immunofluorescence was carried out to characterize NG2-positive cells of vessel walls. GD3 and NG2 expression was investigated in GCSCs and PCSCs whose tumorigenicity was also evaluated in Scid/bg mice. GD3 and NG2 expression was higher in tumor tissue than in BAT. NG2 decreased as the distance from tumor margin increased, regardless of the tumor cell presence, whereas GD3 correlated with neoplastic infiltration. In BAT, NG2 was coexpressed with a-smooth muscle actin (a-SMA) in pericytes and with nestin in the endothelium. Higher levels of NG2 mRNA and protein were found in GCSCs while GD3 synthase was expressed at similar levels in the 2 CSC populations. PCSCs had lower tumorigenicity than GCSCs. These data suggest the possible involvement of GD3 and NG2 in pre/pro-tumorigenic events occurring in the complex microenvironment of the tissue surrounding GBM.


Subject(s)
Antigens/metabolism , Brain Chemistry/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Gangliosides/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Proteoglycans/metabolism , Stem Cells/metabolism , Actins/biosynthesis , Actins/genetics , Adult , Aged , Animals , Antigens/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Gangliosides/genetics , Humans , Immunohistochemistry , Karnofsky Performance Status , Male , Mice , Mice, SCID , Middle Aged , Nestin/biosynthesis , Proteoglycans/genetics , Sialyltransferases/biosynthesis , Sialyltransferases/genetics , Young Adult
7.
Anticancer Res ; 35(4): 1881-916, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25862842

ABSTRACT

AIM: to identify biological interactions between proliferating fibroblasts and HeLa cells in vitro. MATERIALS AND METHODS: Fibroblasts were isolated from both normal and tumour human tissues. Coverslip co-cultures of HeLa and fibroblasts in various ratios with medium replacement every 48 h were studied using fixed cell staining with dyes such as Giemsa and silver staining, with immunochemistry for Ki-67 and E-cadherin, with dihydrofolate reductase (DHFR) enzyme reaction, as well as live cell staining for non-specific esterases and lipids. Other techniques included carmine cell labeling, autoradiography and apoptosis assessment. RESULTS: Under conditions of feeding and cell: cell ratios allowing parallel growth of human fibroblasts and HeLa cells, co-cultured for up to 20 days, a series of phenomena occur consecutively: profound affinity between the two cell types and exchange of small molecules; encircling of the HeLa colonies by the fibroblasts and enhanced growth of both cell types at their contact areas; expression of carbonic anhydrase in both cell types and high expression of non-specific esterases and cytoplasmic argyrophilia in the surrounding fibroblasts; intense production and secretion of lipid droplets by the surrounding fibroblasts; development of a complex net of argyrophilic projections of the fibroblasts; E-cadherin expression in the HeLa cells; from the 10th day onwards, an increasing detachment of batches of HeLa cells at the peripheries of colonies and appearance of areas with many multi-nucleated and apoptotic HeLa cells, and small HeLa fragments; from the 17th day, appearance of fibroblasts blocked at the G2-M phase. Co-cultures at approximately 17-20 days display a cell-cell fight with foci of (a) sparse growth of both cell types, (b) overgrowth of the fibroblasts and (c) regrowth of HeLa in small colonies. These results indicate that during their interaction with HeLa cells in vitro, proliferating fibroblasts can be activated against HeLa. This type of activation is not observed if fibroblast proliferation is blocked by contact inhibition of growth at confluency, or by omitting replacement of the nutrient medium. CONCLUSION: The present observations show that: (a) interaction between proliferating fibroblasts and HeLa cells in vitro drastically influences each other's protein expression, growth pattern, chromatin features and survival; (b) these functions depend on the fibroblast/HeLa ratio, cell topology (cell-cell contact and the architectural pattern developed during co-culture) and frequent medium change, as prerequisites for fibroblast proliferation; (c) this co-culture model is useful in the study of the complex processes within the tumour microenvironment, as well as the in vitro reproduction and display of several phenomena conventionally seen in tumour cytological sections, such as desmoplasia, apoptosis, nuclear abnormalities; and (d) overgrown fibroblasts adhering to the boundaries of HeLa colonies produce and secrete lipid droplets.


Subject(s)
Cell Proliferation/genetics , In Vitro Techniques , Tumor Microenvironment/genetics , Cell Communication/genetics , Cell Survival/genetics , Chromatin/genetics , Coculture Techniques , Fibroblasts/metabolism , Fibroblasts/pathology , HeLa Cells , Humans , Stromal Cells/pathology
8.
PLoS One ; 8(3): e57145, 2013.
Article in English | MEDLINE | ID: mdl-23472076

ABSTRACT

The gene expression pattern of glioblastoma (GBM) is well documented but the expression profile of brain adjacent to tumor is not yet analysed. This may help to understand the oncogenic pathway of GBM development. We have established the genome-wide expression profiles of samples isolated from GBM tumor mass, white matter adjacent to tumor (apparently free of tumor cells), and white matter controls by using the Affymetrix HG-U133 arrays. Array-CGH (aCGH) was also performed to detect genomic alterations. Among genes dysregulated in peritumoral white matter, 15 were over-expressed, while 42 were down-regulated when compared to white matter controls. A similar expression profile was detected in GBM cells. Growth, proliferation and cell motility/adhesion-associated genes were up-regulated while genes involved in neurogenesis were down-regulated. Furthermore, several tumor suppressor genes along with the KLRC1 (a member of natural killer receptor) were also down-regulated in the peritumoral brain tissue. Several mosaic genomic lesions were detected by aCGH, mostly in tumor samples and several GBM-associated mosaic genomic lesions were also present in the peritumoral brain tissue, with a similar mosaicism pattern. Our data could be explained by a dilution of genes expressed from tumor cells infiltrating the peritumour tissue. Alternatively, these findings could be substained by a relevant amount of "apparently normal" cells presenting a gene profile compatible with a precancerous state or even "quiescent" cancer cells. Otherwise, the recurrent tumor may arise from both infiltrating tumor cells and from an interaction and recruitment of apparently normal cells in the peritumor tissue by infiltrating tumor cells.


Subject(s)
Brain Neoplasms/metabolism , Brain/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Transcriptome , Brain/metabolism , Brain Neoplasms/genetics , Cell Adhesion , Cell Movement , Cell Proliferation , Cluster Analysis , Comparative Genomic Hybridization , Genome-Wide Association Study , Glioblastoma/genetics , Humans , Neurons/metabolism , Oligonucleotide Array Sequence Analysis
9.
PLoS One ; 8(1): e52530, 2013.
Article in English | MEDLINE | ID: mdl-23326338

ABSTRACT

High cell-surface GnRH receptor (GnRH-R) levels have been shown to have a major influence on the extent of GnRH agonist-mediated tumor growth inhibition. The ability of the GnRH agonist leuprorelin acetate (LA) to induce a post-transcriptional upregulation of GnRH-R at the plasma membrane of androgen-sensitive (LNCaP) and -insensitive (PC-3) prostate cancer (PCa) cells has been previously demonstrated by Western blotting. Here we performed single molecule force spectroscopy by using Atomic Force Microscopy (AFM), which has proven to be a powerful tool allowing for investigation of living cell surface biological features, such as the so far unclear GnRH agonist/receptor interaction. Thus, in the hormone-insensitive PC-3 cells, we characterized the strength of the LA-receptor binding, and the amount and distribution of the functional receptor molecules on the cell surface. The effect of a long and continuous treatment (up to 30 days) with the agonist (10(-11) and 10(-6) M) on the same parameters was also investigated. A GnRH-R increase was observed, reaching the maximum (∼80%) after 30 days of treatment with the highest dose of LA (10(-6) M). The analogue-induced increase in GnRH-R was also demonstrated by Western blotting. In addition, two different receptor bound strengths were detected by AFM, which suggests the existence of two GnRH-R classes. A homogeneous distribution of the unbinding events has been found on untreated and treated PC-3 cell surfaces. The persistence of high receptor levels at the membrane of these living cells may warrant the maintenance of the response to LA also in androgen-unresponsive PCa. Moreover, the determination of ligand/receptor bond strength could shed light on the poorly understood event of LA/GnRH-R interaction and/or address structural/chemical agonist optimizations.


Subject(s)
Leuprolide/metabolism , Microscopy, Atomic Force/methods , Receptors, LHRH/metabolism , Antineoplastic Agents, Hormonal/metabolism , Antineoplastic Agents, Hormonal/pharmacology , Binding Sites , Binding, Competitive , Blotting, Western , Cell Line, Tumor , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , HEK293 Cells , Humans , Leuprolide/pharmacology , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, LHRH/agonists , Receptors, LHRH/genetics
10.
Clin Neurol Neurosurg ; 115(7): 883-6, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22959214

ABSTRACT

OBJECTIVE: Treatment of glioblastoma recurrence can have a palliative aim, after considering risks and potential benefits. The aim of this study is to verify the impact of surgery and of palliative adjuvant treatments on survival after recurrence. METHODS: From January 2002 to June 2008, we treated 76 consecutive patients with recurrent glioblastoma. Treatment was: 1-surgery alone--17 patients; 2-adjuvant-therapy alone--24 patients; 3-surgery and adjuvant therapy--16 patients; no treatment--19 patients. The impact on median overall-survival (OS-time between recurrence and death/last follow-up) of age, Karnofsky performance scale (KPS), resection extent and adjuvant treatment scheme (Temozolomide alone vs low-dose fractionated radiotherapy vs others) was determined. Survival curves were obtained through the Kaplan-Meier method. Cox proportional-hazards was used for multivariate analyses. Significance was set at p<0.05. RESULTS: Median OS was 7 months. At univariate analysis, patients with a KPS≥70 had a longer OS (9 months vs 5 months--p<0.0001). OS was 6 months for patients treated with surgery alone, 5 months for patients that received no treatment, 8 months for patients treated with chemotherapy alone, 14 months for patients treated with surgery and adjuvant therapy--p=0.01. Patients with a KPS<70 were significantly at risk for death - HR 2.8 - p=0.001. Subgroup analysis showed no significant differences between patients receiving gross total or partial tumor resection and among patients receiving different adjuvant therapy schemes. Major surgical morbidity at tumor recurrence occurred in 16 out of 33 patients (48%). CONCLUSION: It is fundamental, before deciding to operate patients for recurrence, to carefully consider the impact of surgical morbidity on outcome.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Adult , Aged , Aged, 80 and over , Brain Neoplasms/diagnostic imaging , Chemoradiotherapy, Adjuvant , Combined Modality Therapy , Confidence Intervals , Data Interpretation, Statistical , Female , Follow-Up Studies , Glioblastoma/surgery , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local , Neurosurgical Procedures/methods , Palliative Care , Regression Analysis , Reoperation , Survival Analysis , Ultrasonography
11.
PLoS One ; 7(12): e50804, 2012.
Article in English | MEDLINE | ID: mdl-23251387

ABSTRACT

Interactions occurring between malignant cells and the stromal microenvironment heavily influence tumor progression. We investigated whether this cross-talk affects some molecular and functional aspects specifically correlated with the invasive phenotype of breast tumor cells (i.e. adhesion molecule expression, membrane fluidity, migration) by co-culturing mammary cancer cells exhibiting different degrees of metastatic potential (MDA-MB-231>MCF-7) with fibroblasts isolated from breast healthy skin (normal fibroblasts, NFs) or from breast tumor stroma (cancer-associated fibroblasts, CAFs) in 2D or 3D (nodules) cultures. Confocal immunofluorescence analysis of the epithelial adhesion molecule E-cadherin on frozen nodule sections demonstrated that NFs and CAFs, respectively, induced or inhibited its expression in MCF-7 cells. An increase in the mesenchymal adhesion protein N-cadherin was observed in CAFs, but not in NFs, as a result of the interaction with both kinds of cancer cells. CAFs, in turn, promoted N-cadherin up-regulation in MDA-MB-231 cells and its de novo expression in MCF-7 cells. Beyond promotion of "cadherin switching", another sign of the CAF-triggered epithelial-mesenchymal transition (EMT) was the induction of vimentin expression in MCF-7 cells. Plasma membrane labeling of monolayer cultures with the fluorescent probe Laurdan showed an enhancement of the membrane fluidity in cancer cells co-cultured with NFs or CAFs. An increase in lipid packing density of fibroblast membranes was promoted by MCF-7 cells. Time-lapsed cell tracking analysis of mammary cancer cells co-cultured with NFs or CAFs revealed an enhancement of tumor cell migration velocity, even with a marked increase in the directness induced by CAFs.Our results demonstrate a reciprocal influence of mammary cancer and fibroblasts on various adhesiveness/invasiveness features. Notably, CAFs' ability to promote EMT, reduction of cell adhesion, increase in membrane fluidity, and migration velocity and directness in mammary cancer cells can be viewed as an overall progression- and invasion-promoting effect.


Subject(s)
Breast Neoplasms/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Cell Movement/physiology , Epithelial Cells/metabolism , Stromal Cells/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Epithelial Cells/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Stromal Cells/pathology , Tumor Cells, Cultured
12.
Autoimmun Rev ; 10(10): 624-30, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21545850

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune disease characterized by excessive extracellular matrix (ECM) deposition in the skin and other visceral organs and it is associated with immune activation characterized by autoantibody production, release of various cytokines and T-lymphocyte activation. Several recent lines of evidence in animal models and in SSc patients indicate a potential role for B cells in the SSc. B cells have arisen as a possible player in tissue fibrosis in some experimental models and, since IL-6 produced by B cells, along with TGF-ß, may induce matrix synthesis and less collagen degradation, targeting B cells could be one way to reduce ECM deposition and reduce the inflammatory background. Both SSc patients and tight-skin mice, a genetic model of SSc, have intrinsic B-cell abnormalities characterized by chronic B-cell activation. SSc patients present an increased number of naïve B cells and an activation of memory B cells, despite a reduction in their number. B cells from SSc patients exhibit increased expression of CD19. Remarkably, CD19 loss or B-cell depletion using antimouse CD20 antibody suppresses the development of skin hyperplasia and autoimmunity in tight-skin mice. Additionally, recent studies revealed a possible beneficial effect of anti-human CD20 antibody (Rituximab) therapy on skin fibrosis and lung involvement in SSc patients. These studies reported also the safety of Rituximab in SSc patients. All these findings suggest a possible role of antiCD20 treatment in SSc patients.


Subject(s)
B-Lymphocytes/immunology , Scleroderma, Systemic/drug therapy , Scleroderma, Systemic/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , B-Lymphocytes/drug effects , Clinical Trials as Topic , Disease Models, Animal , Fibrosis , Humans , Lymphocyte Activation/drug effects , Lymphocyte Depletion , Mice , Mice, Mutant Strains , Molecular Targeted Therapy , Rituximab , Scleroderma, Systemic/genetics , Scleroderma, Systemic/physiopathology
13.
Int J Oncol ; 38(6): 1501-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21479359

ABSTRACT

Reduced cell-cell adhesion allows malignant epithelial cells to invade the basal membrane and penetrate the stroma. This implies the potential of the cells to escape from the primary tumor as well as spreading ability. Herein, we investigated the effects of leuprorelin acetate (LA), a GnRH agonistic analogue, alone or in combination with dihydrotestosterone (DHT), on the expression of molecules involved in cell adhesion (E-cadherin, N-cadherin, α-, ß- and γ-catenin) or in migration/invasion (c-met, CD44v6 and caveolin-1) in androgen-sensitive (LNCaP) and -insensitive (PC-3) prostate cancer (CaP) cells. We demonstrated by immunoblotting that, in LNCaP cells, molecules present in the adherens junctions (E-cadherin, α-, ß- and γ-catenin) were expressed, while α-catenin was absent in PC-3 cells which expressed N-cadherin and c-met. In LNCaP cells, no changes in E-cadherin levels were produced by 10(-9) M DHT while LA (10(-11) or 10(-6) M) up-regulated the protein level (up to 26-30% after 48 h). In the same cells, ß- and γ-catenin expression was enhanced either by DHT (24 and 20%, respectively) or LA (up to 18 and 40%, respectively), while α-catenin was not affected. Antagonistic effects were consistently observed between DHT and LA when the two hormones were jointly administered to the cells. Consistent results were obtained by immunocytochemistry. Co-immunoprecipitation analysis, used to verify the integrity of the LNCaP cell adhesion complex, demonstrated the association of E-cadherin with catenins. In PC-3 cells, adhesion molecule expression, analyzed by immunoblotting, was unaffected by LA, while a down-regulation of c-met (up to 28%) was observed after 24 h of treatment but which did not hold up over time (48-144 h). Our findings demonstrate the efficacy of LA in upregulating E-cadherin, ß- and γ-catenin in LNCaP cells. This effect, that may be considered as another aspect of the direct antitumor activity of the GnRH analogue in hormone-dependent CaP cells, may contribute to maintenance/restoration of the normal tissue architecture counteracting the tumor cell spreading tendency.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cell Adhesion Molecules/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Leuprolide/pharmacology , Cell Line, Tumor , Dihydrotestosterone/pharmacology , Humans , Immunohistochemistry , Male , Prostatic Neoplasms
14.
Int J Oncol ; 38(1): 41-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21109924

ABSTRACT

Angiogenesis in the peritumor tissue of glioblastoma (GBM) is still an open field of research. This study investigates neovascularization in the tumor surrounding areas by examining CD105 and nestin expression along with microvessel density (MVD) with the aim of establishing their possible prognostic significance. Angiogenesis was also confirmed by investigating, in vessel walls, the presence of pericytes, which are multipotent stem cells, expressing α-smooth muscle actin (α-SMA). In our study, including 40 GBM patients, tissue samples were obtained from tumors (first area) and white matter at a distance <1 cm (second area) and between 1 and 3.5 cm (third area) from the tumor margin. CD105 and nestin were detected by immunohistochemistry in hyperplastic endothelium of GBM and peritumor tissue, and occasionally coexpressed or colocalized. Pericytes encircling hyperplastic endothelium were evident in all three areas. Univariate analysis revealed that patients with a CD105-MVD value ≥8 in the third area have a significantly shorter survival time and Cox analysis indicated an about 3.5-fold increase in death risk in the same patients. These results demonstrate that a tumor neoangiogenesis occurs in GBM peritumor tissue with intimate involvement of pericytes. CD105-MVD in the area located at a greater distance from the tumor margin carries prognostic significance.


Subject(s)
Antigens, CD/biosynthesis , Glioblastoma/blood supply , Intermediate Filament Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Receptors, Cell Surface/biosynthesis , Supratentorial Neoplasms/blood supply , Actins/metabolism , Adult , Aged , Endoglin , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Immunohistochemistry , Male , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nestin , Supratentorial Neoplasms/metabolism , Supratentorial Neoplasms/pathology
15.
Arthritis Res Ther ; 12(2): R54, 2010.
Article in English | MEDLINE | ID: mdl-20338043

ABSTRACT

INTRODUCTION: An over-expression of CD19 has been shown in B cells of systemic sclerosis (SSc) and B cells are thought to contribute to the induction of skin fibrosis in the tight skin mouse model. The aim was to define the outcome on safety and the change in skin score after rituximab therapy in SSc patients and to correlate the clinical characteristics with the levels of interleukin (IL)-6 and with the immune cell infiltrate detected by immunohistochemistry. METHODS: Nine patients with SSc with mean age 40.9 +/- 11.1 years were treated with anti-CD20, 1 g at time 0 and after 14 days. Skin biopsy was performed at baseline and during the follow-up. B-cell activating factor (BAFF) and IL-6 levels were also determined at the follow-up times. RESULTS: After 6 months patients presented a median decrease of the skin score of 43.3% (range 21.1-64.0%), and a decrease in disease activity index and disease severity index. IL-6 levels decreased permanently during the follow up. After treatment, a complete depletion of peripheral blood B cells was observed in all but 2 patients. Only 3 patients presented CD20 positive cells in the biopsy of the involved skin at baseline. CONCLUSIONS: Anti-CD20 treatment has been well tolerated and SSc patients experienced an improvement of the skin score and of clinical symptoms. The clear fall in IL-6 levels could contribute to the skin fibrosis improvement, while the presence of B cells in the skin seems to be irrelevant with respect to the outcome after B cell depletion. TRIAL REGISTRATION: ISRCTN77554566.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, CD20/immunology , B-Lymphocytes/drug effects , Immunologic Factors/therapeutic use , Scleroderma, Systemic/drug therapy , Skin/drug effects , Adult , Antibody-Dependent Cell Cytotoxicity , B-Cell Activating Factor/metabolism , B-Lymphocytes/pathology , Female , Health Status , Humans , Interleukin-6/metabolism , Lymphocyte Depletion , Male , Middle Aged , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Severity of Illness Index , Skin/metabolism , Skin/pathology , Young Adult
16.
Endocrine ; 36(1): 87-97, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19399647

ABSTRACT

GnRH receptors (GnRH-R) have been found in various malignancies, including prostate cancer (PCa). They mediate the direct antitumor effects of GnRH analogs. Nevertheless, few reports concern drug-induced modulation of GnRH-R levels. In this study, we investigated GnRH-R expression in androgen-sensitive (LNCaP) and -insensitive (PC-3) PCa cells treated for 4 and 6 days with a GnRH agonist (Leuprorelin acetate, LA, 10(-11) or 10(-6) M), Dihydrotestosterone (DHT, 10(-9) M), Cyproterone acetate (CA, 10(-7) M), and Epidermal growth factor (EGF, 10 ng/ml), either alone or combined. The RT-PCR analysis showed no variation in GnRH-R mRNA levels of both treated LNCaP and PC-3 cells. On the contrary, immunoblotting indicated that in LNCaP and PC-3 cells, LA upregulated membrane GnRH-R expression (up to 92%). In androgen-sensitive cells, DHT induced a GnRH-R increase (up to 119%) always comparable to that occurring in the presence of CA. GnRH-R upregulation by LA/DHT or CA/DHT association was similar to that promoted by the single agents. In PC-3 cells, EGF upregulated GnRH-R (up to 110%). A prolonged treatment (for 12 days) determined a greater EGF-induced increase in GnRH-R levels (142%). Lower (or no) receptor enhancement occurred when LA and EGF were associated. Our findings indicate that LA post-transcriptionally upregulates its own membrane receptor in androgen-sensitive and -insensitive PCa cells, counteracting the receptor enhancement produced by DHT and EGF. The effects, obtained with a relatively long and continuous treatment, may have implications in the choice of therapy modality with GnRH analogs and may render the receptor a novel therapeutic target, particularly in hormone-refractory PCa.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Leuprolide/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Receptors, LHRH/genetics , Androgen Antagonists/pharmacology , Androgens/pharmacology , Blotting, Western , Cell Line, Tumor , Cyproterone Acetate/pharmacology , Dihydrotestosterone/pharmacology , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/pathology , RNA, Messenger/metabolism , Receptors, LHRH/metabolism , Up-Regulation/drug effects
17.
Cancer ; 113(4): 841-6, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18618580

ABSTRACT

BACKGROUND: After surgical resection, the residual, invasive glioblastoma (GBM) cells give rise to a recurrent tumor, which, in 96% of patients, arises adjacent to the resection margin. METHODS: In this study, the authors prospectively enrolled 25 patients with GBM who underwent gross total resection followed by adjuvant radiochemotherapy (with temozolomide). Tumor removal was achieved with resection margins that included the neighboring, apparently normal tissue (between 1 cm and 2 cm from the tumor border [B area]) and the tumor. RESULTS: Patients who had an absence of tumor cells in the neighboring, apparently normal white matter (B area) had better survival than patients who had the presence of tumor cells in the B area (21 months vs 12 months). This difference was statistically significant in univariate analysis (P = .005) and in multivariate analysis (P = .01). CONCLUSIONS: Aggressive tumor removal may improve survival, but the current results indicated that biologic commitment of 'penumbra' cells appear to be the most relevant factor for tumor recurrence and accounts for the fatal outcome of the disease.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Aged , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Female , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Glioblastoma/surgery , Humans , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Survival Analysis , Temozolomide , Treatment Outcome
18.
Clin Cancer Res ; 13(23): 6970-7, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056172

ABSTRACT

PURPOSE: It has been hypothesized that brain tumors are derived from stem cell or transiently dividing precursor transformation. Furthermore, c-Jun NH(2)-terminal kinases (JNKs) have been involved in gliomagenesis. This study analyzes stem cell marker nestin and JNK expression in glioblastoma multiforme (GBM) and peritumor tissue and assesses their possible prognostic implications. EXPERIMENTAL DESIGN: Nestin and both total JNK (tJNK) and phosphorylated JNK (pJNK) expression was investigated by immunohistochemistry in 20 GBMs. Samples were derived from tumors (first area), from tissues at a distance <1 cm (second area), and between 1 and 3.5 cm (third area) from the macroscopic tumor border. The relationships between patients' age, Karnofsky performance status, gender, protein expression, and survival were analyzed. RESULTS: Nestin cytoplasmic immunoreactivity was observed in the majority of cells in tumor but infrequently in peritumor areas. tJNK, observed in the nucleus and cytoplasm, was widely expressed in the three areas; pJNK, mostly located in the nuclei, was found in a variable percentage of cells in the tumor and peritumor tissue. Nestin and JNK expression in peritumor areas was independent of the presence of neoplastic cells. Univariate analysis indicated that survival was longer (19 versus 12 months; P = 0.01) for patients whose pJNK/nestin and (pJNK/tJNK)/nestin ratios in the second area were > or =2.619 and > or =0.026, respectively. The same variables showed an independent prognostic value in multivariate analysis. CONCLUSIONS: Nestin and JNK expression indicates that peritumor tissue, independently of the presence of neoplastic cells, may present signs of transformation. Moreover, pJNK/nestin and (pJNK/tJNK)/nestin ratios in that tissue seem to have some prognostic implications in GBM patients.


Subject(s)
Glioblastoma/metabolism , Glioblastoma/pathology , Intermediate Filament Proteins/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , Nerve Tissue Proteins/biosynthesis , Supratentorial Neoplasms/metabolism , Supratentorial Neoplasms/pathology , Adult , Aged , Enzyme Activation , Female , Glioblastoma/enzymology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Nestin , Prognosis , Stem Cells/metabolism , Stem Cells/pathology , Supratentorial Neoplasms/enzymology
19.
Int J Oncol ; 30(6): 1333-42, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17487353

ABSTRACT

Anomalies of growth factor signaling have been reported in malignant human gliomas. The extracellular signal-regulated kinases (ERKs) play a crucial role in transducing growth factor signals to the nucleus and are involved in a wide range of biological responses, including cell proliferation, differentiation and motility. ERK1/2 is expressed and activated in glioblastoma multiforme (GBM). However, no information is available in literature concerning the presence and activity of ERK1/2 in the peritumor tissue. In the present study, we evaluated by immunohistochemistry total and phosphorylated (t and p) ERK1/2 expression in 31 cases of primary GBM and in tissue surrounding the enhanced lesion at different distances up to 3.5 cm from the tumor margin. Total ERK1/2 was, as expected, uniformly expressed not only in GBM but in the areas around the tumor also, which showed higher levels of immunolabeling. ERK1/2 activation was observed in GBM as well as in peritumor tissue, with no statistical difference in the level of the enzymatic activities. In particular, in the peritumor tissue pERK1/2 was present independently of neoplastic cells not only in reactive astrocytes, but in apparently normal glial cells also. These results indicate that ERK1/2 pathway may participate in GBM growth and progression. In addition, they strongly suggest that ERK1/2 stimulation may be linked not only to tissue reactivity to tumor invasion, but also to cell motility or represent per se a sign of transformation. Finally, our findings highlight the meaning of the extension of neoplasm fingers beyond the outer margin of GBM. Patients with neoplastic cells at <10% or without neoplastic cells in peritumor areas showed a higher survival time compared with those with neoplastic elements at > or = 10%. In addition, a percentage > or = 10 of neoplastic elements in peritumor tissue was associated with an approximately 4-fold increased death risk.


Subject(s)
Brain Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glioblastoma/metabolism , Adult , Aged , Brain Neoplasms/mortality , Female , Glioblastoma/mortality , Humans , Immunohistochemistry , Male , Middle Aged , ROC Curve , Sensitivity and Specificity , Survival Analysis
20.
Int J Oncol ; 29(1): 237-47, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16773205

ABSTRACT

The mechanisms by which a GnRH analogue, leuprorelin acetate (LA), antagonizes the mitogenic effect of dihydrotestosterone (DHT) or epidermal growth factor (EGF) in prostate cancer cells is poorly understood. The mitogen-activated protein kinase system has a central role in growth regulation and, for this reason, we investigated the involvement of the extracellular signal-regulated kinase (ERK1/2) pathway in the response of both androgen-sensitive (LNCaP) and -insensitive (PC-3) prostate cancer cells to LA alone or combined with EGF or DHT. The evaluation of ERK activation was performed by using Western blot analysis and immunocytochemistry. EGF specifically induced ERK1/2 activity in both models and this effect was counteracted by an inhibitor of EGF-receptor phosphorylation. The addition of LA produced an appreciable reduction of ERK phosphorylation promoted by EGF in LNCaP cells, while it generally determined an increase in ERK activity in androgen-unresponsive PC-3 cells. The slight ERK activation induced by DHT in LNCaP cells was counteracted by LA and this effect was evident only by immunocytochemistry. Our findings suggest that the antiproliferative effect of LA in prostate cancer cells stimulated by hormones and growth factors may be, at least in part, mediated by the reduction of ERK1/2 activation in LNCaP cells and linked to the unexpected increase of this activity produced by the analogue in PC-3 cells.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Leuprolide/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasms, Hormone-Dependent/enzymology , Prostatic Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Dihydrotestosterone/pharmacology , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Humans , Male , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Quinazolines , Time Factors , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL