Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
2.
Br J Cancer ; 117(7): 994-1006, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28772282

ABSTRACT

BACKGROUND: Besides its role as oxygen transporter, recent findings suggest that haemoglobin beta (HBB) may have roles in other contexts. METHODS: We evaluated the impact of HBB expression in primary human breast cancers, and in breast cancer cell lines overexpressing HBB by in vitro and in vivo studies. Publicly available microarray databases were used to perform multivariate survival analyses. RESULTS: A significantly higher expression of HBB was observed in invasive carcinoma histotypes vs in situ counterparts, along with a positive correlation between HBB and the Ki67 proliferation marker. HBB-overexpressing breast cancer cells migrate and invade more, show HIF-1α upregulation and their conditioned media enhances angiogenesis. Blocking the oxygen-binding site of HBB reverts the increase of migration and HIF-1α upregulation observed in HBB-overexpressing breast cancer cells. Orthotopically implanted MDA-MB-231 overexpressing HBB (MDA-HBB) generated tumours with faster growth rate and increased neoangiogenesis. Moreover, local recurrence and visceral metastases were observed only in MDA-HBB-implanted mice. Similar results were observed with 4T1 mouse breast cancer cells. Finally, bioinformatics analyses of public data sets correlated high HBB expression with lower overall survival. CONCLUSIONS: HBB expression increases breast cancer cells aggressiveness and associates with poor prognosis, pointing to HBB as a novel biomarker for breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Lobular/metabolism , Neoplasm Recurrence, Local/metabolism , Neovascularization, Pathologic/metabolism , beta-Globins/metabolism , Animals , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/secondary , Carcinoma, Intraductal, Noninfiltrating/chemistry , Carcinoma, Lobular/chemistry , Carcinoma, Lobular/secondary , Cell Line, Tumor , Cell Movement , Computational Biology , Female , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ki-67 Antigen/analysis , Lymph Nodes/chemistry , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Oxidative Stress , Survival Rate , Tissue Array Analysis , beta-Globins/analysis , beta-Globins/genetics
3.
Oncotarget ; 8(69): 113792-113806, 2017 Dec 26.
Article in English | MEDLINE | ID: mdl-29371946

ABSTRACT

Despite the growing body of knowledge showing that testosterone (T) may not significantly affect tumor progression in hypogonadal patients treated for prostate cancer (Pca), the use of this hormone in this population still remains controversial. The effects of continuous or pulsed T stimulation were tested in vitro and in vivo on androgen-sensitive Pca cell lines in order to assess the differential biological properties of these two treatment modalities. Pulsed T treatment resulted in a greater inhibition than continuous T supplementation of tumor growth in vitro and in vivo. The effects of pulsed T treatment on tumor growth inhibition, G0/G1 cell cycle arrest, and tumor senescence was more pronounced than those obtained upon continuous T treatments. Mechanistic studies revealed that G0/G1 arrest and tumor senescence upon pulsed T treatment were associated with a marked decrease in cyclin D1, c-Myc and SKp2, CDK4 and p-Rb levels and upregulation of p27 and p-ERK1/2. Pulsed, but not continuous, T supplementation decreased the expression levels of AR, p-ARser81 and CDK1 in both cellular models. The in vitro results were confirmed in an in vivo xenografts, providing evidence of a greater inhibitory activity of pulsed supraphysiological T supplementation than continuous treatment, both in terms of tumor volume and decreased AR, p-ARser81, PSA and CDK1 staining. The rapid cycling from hypogonadal to physiological or supra-physiological T intraprostatic concentrations results in cytostatic and senescence effects in preclinical models of androgen-sensitive Pca. Our preclinical evidence provides relevant new insights in the biology of Pca response to pulsed T supplementation.

4.
Psychiatry Res ; 244: 165-70, 2016 Oct 30.
Article in English | MEDLINE | ID: mdl-27479108

ABSTRACT

Clinical correlates of plasma Brain-Derived Neurotrophic Factor (BDNF) have been investigated in a clinical population with Post Traumatic Stress Disorder (PTSD) symptoms and healthy control subjects who survived to the L'Aquila 2009 earthquake. Twenty-six outpatients and 14 control subjects were recruited. Assessments included: Structured Clinical Interview for DSM-IV Axis-I disorders Patient Version, Trauma and Loss Spectrum-Self Report (TALS-SR) for post-traumatic spectrum symptoms. Thirteen patients were diagnosed as Full PTSD and 13 as Partial PTSD. The subjects with full-blown PTSD showed lower BDNF level than subjects with partial PTSD and controls. Different relationship patterns of BDNF with post-traumatic stress spectrum symptoms have been reported in the three samples. Our findings add more insight on the mechanisms regulating BDNF levels in response to stress and further proofs of the utility of the distinction of PTSD into full and partial categories.


Subject(s)
Brain-Derived Neurotrophic Factor/blood , Disasters , Earthquakes , Stress Disorders, Post-Traumatic/blood , Adult , Case-Control Studies , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , Italy , Male , Middle Aged , Self Report , Severity of Illness Index , Stress Disorders, Post-Traumatic/psychology
5.
Tumour Biol ; 37(9): 12743-12753, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27448307

ABSTRACT

Glioblastoma has one of the highest mortality rates among cancers, and it is the most common and malignant form of brain cancer. Among the typical features of glioblastoma tumors, there is an aberrant vascularization: all gliomas are among the most vascularized/angiogenic tumors. In recent years, it has become clear that glioblastoma cells can secrete extracellular vesicles which are spherical and membrane-enclosed particles released, in vitro or in vivo, by both normal and tumor cells; they are involved in the regulation of both physiological and pathological processes; among the latter, cancer is the most widely studied. Extracellular vesicles from tumor cells convey messages to other tumor cells, but also to normal stromal cells in order to create a microenvironment that supports cancer growth and progression and are implicated in drug resistance, escape from immunosurveillance and from apoptosis, as well as in metastasis formation; they are also involved in angiogenesis stimulation, inducing endothelial cells proliferation, and other pro-angiogenic activities. To this aim, the present paper assesses in detail the extracellular vesicles phenomenon in the human glioblastoma cell line U251 and evaluates extracellular vesicles ability to promote the processes required to achieve the formation of new blood vessels in human brain microvascular endothelial cells, highlighting that they stimulate proliferation, motility, and tube formation in a dose-response manner. Moreover, a molecular characterization shows that extracellular vesicles are fully equipped for angiogenesis stimulation in terms of proteolytic enzymes (gelatinases and plasminogen activators), pro-angiogenic growth factors (VEGF and TGFß), and the promoting-angiogenic CXCR4 chemokine receptor.


Subject(s)
Culture Media, Conditioned/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Neovascularization, Pathologic/metabolism , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/physiopathology , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Exosomes/metabolism , Exosomes/ultrastructure , Extracellular Vesicles/ultrastructure , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Neovascularization, Pathologic/physiopathology , Receptors, CXCR4/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Growth Factors ; 34(3-4): 87-96, 2016 08.
Article in English | MEDLINE | ID: mdl-27362575

ABSTRACT

Recent studies suggest that adipose-derived stem cells (ASCs) play a role in tissue remodeling through the release of cytokines and growth factors. We compared the secreted cytokine profile of hypoxia-conditioned ASCs (hASCs) with normoxic ASCs (nASCs) and we analyzed the effect of ASCs conditioned medium (CM) on endothelial cells. We found that hypoxia induced a transient upregulation of VEGF in ASCs and a notable and enduring upregulation of leptin mRNA expression 30-fold greater than control after 24 h and up to 60-fold greater than control at day 7. CM from hASC stimulated EC tube formation to a significantly greater extent than CM from nASC. This might be due to leptin-secreted factor. Indeed, exogenous leptin stimulated the expression of HIF2-α, but not HIF1-α, and upregulated the expression of Flt-1 and Tie-1 proangiogenic receptors. In conclusion, hASCs may be particularly efficient in sustaining angiogenesis through the release of leptin.


Subject(s)
Adipose Tissue/cytology , Adult Stem Cells/metabolism , Endothelial Cells/cytology , Leptin/metabolism , Neovascularization, Physiologic , Adipose Tissue/metabolism , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leptin/genetics , Leptin/pharmacology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Oncol Rep ; 36(1): 125-30, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27222100

ABSTRACT

The high affinity nerve growth factor (NGF) NGF receptor, p75NTR, is a member of the tumor necrosis factor (TNF) receptor superfamily that shares a conserved intracellular death domain capable of inducing apoptosis and suppressing growth in prostate epithelial cells. Expression of this receptor is lost as prostate cancer progresses and is minimal in established prostate cancer cell lines. We aimed to verify the role of p75NTR in the azacitidine-mediated antitumor effects on 22Rv1 and PC3 androgen-independent prostate cancer cells. In the present study, we reported that the antiproliferative and pro-apoptotic effects of 5-azacytidine (azacitidine) were more marked in the presence of physiological concentrations of NGF and were reduced when a blocking p75NTR antibody or the selective p75NTR inhibitor, Ro 08-2750, were used. Azacitidine increased the expression of p75NTR without interfering with the expression of the low affinity NGF receptor TrkA and induced caspase 9-dependent caspase 3 activity. Taken together, our results suggest that the NGF network could be a candidate for future pharmacological manipulation in aggressive prostate cancer.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Azacitidine/pharmacology , Cell Proliferation/drug effects , Nerve Tissue Proteins/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Receptors, Nerve Growth Factor/metabolism , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Flavins , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Nerve Tissue Proteins/antagonists & inhibitors , Pteridines/pharmacology , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Transplantation, Heterologous
8.
Mol Cell Endocrinol ; 426: 61-72, 2016 May 05.
Article in English | MEDLINE | ID: mdl-26872613

ABSTRACT

PURPOSE: To search for a possible role of Peroxisome Proliferator-Activated Receptor α (PPARα), a molecular partner of the Aryl hydrocarbon receptor Interacting Protein (AIP), in somatotropinomas. METHODS: Tumours from 51 acromegalic patients were characterized for PPARα and AIP expression by immunohistochemistry (IHC) and/or Real Time RT-PCR. Data were analysed according to tumour characteristics and pre-operative treatment with somatostatin analogues (SSA). The effects of fenofibrate were studied in GH3 cells in vitro. RESULTS: PPARα was expressed in most somatotropinomas. A modest relationship was found between PPARα and AIP expression, both being significantly higher in the presence of pre-operative SSA. However, only AIP expression was influenced by the response to treatment. Dual effects of fenofibrate were observed in GH3 cells, consisting of cell growth inhibition and an increase in GH secretion inhibited by octreotide. CONCLUSIONS: PPARα is a new player in somatotropinomas. Potential interactions between PPARα agonists and SSA may deserve further investigation.


Subject(s)
Adenoma/metabolism , Fenofibrate/pharmacology , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , PPAR alpha/metabolism , Adolescent , Adult , Aged , Animals , Apoptosis , Cell Line , Cell Proliferation , Child , Female , Humans , Male , Middle Aged , Pituitary Gland/metabolism , Pituitary Gland/pathology , Rats , Young Adult
10.
BMC Cancer ; 15: 941, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620414

ABSTRACT

BACKGROUND AND AIMS: Increased expression of Chromosome Region Maintenance (CRM-1)/exportin-1 (XPO-1) has been correlated with poor prognosis in several aggressive tumors, making it an interesting therapeutic target. Selective Inhibitor of Nuclear Export (SINE) compounds bind to XPO-1 and block its ability to export cargo proteins. Here, we investigated the effects of a new class of SINE compounds in models of prostate cancer. MATERIAL AND METHODS: We evaluated the expression of XPO-1 in human prostate cancer tissues and cell lines. Next, six SINE (KPT-127, KPT-185, KPT-205, KPT-225, KPT-251 and KPT-330) compounds having different potency with broad-spectrum, tumor-selective cytotoxicity, tolerability and pharmacokinetic profiles were tested in a panel of prostate cancer cells representing distinct differentiation/progression states of disease and genotypes. Two SINE candidates for clinical trials (KPT-251 and KPT-330) were also tested in vivo in three cell models of aggressive prostate cancer engrafted in male nude mice. RESULTS AND CONCLUSIONS: XPO-1 is overexpressed in prostate cancer compared to normal or hyperplastic tissues. Increased XPO-1 expression, mainly in the nuclear compartment, was associated with increased Gleason score and bone metastatic potential supporting the use of SINEs in advanced prostate cancer. SINE compounds inhibited proliferation and promoted apoptosis of tumor cells, but did not affect immortalized non-transformed prostate epithelial cells. Nuclei from SINE treated cells showed increased protein localization of XPO-1, survivin and cyclin D1 followed by degradation of these proteins leading to cell cycle arrest and apoptosis. Oral administration of KPT-251 and KPT-330 in PC3, DU145 and 22rv1 tumor-bearing nude mice reduced tumor cell proliferation, angiogenesis and induced apoptosis. Our results provide supportive evidence for the therapeutic use of SINE compounds in advanced/castration resistant prostate cancers and warrants further clinical investigation.


Subject(s)
Analgesics/administration & dosage , Cyclin D1/metabolism , Hydrazines/administration & dosage , Inhibitor of Apoptosis Proteins/metabolism , Prostatic Neoplasms/drug therapy , Triazoles/administration & dosage , Adult , Analgesics/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydrazines/pharmacology , Male , Mice , Mice, Nude , Neoplasm Grading , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacology , Prostatic Neoplasms/metabolism , Survivin , Triazoles/pharmacology , Up-Regulation , Xenograft Model Antitumor Assays
11.
Transl Androl Urol ; 4(2): 139-47, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26816820

ABSTRACT

The literature suggests that the serum testosterone level required for maximum androgen receptor (AR) binding may be in the range of nanomolar and above this range of concentrations; this sexual hormone may not significantly affect tumour biology. This assumption is supported by clinical studies showing that cell proliferation markers did not change when serum T levels increased after exogenous T treatment in comparison to subjects treated with placebo. However, a considerable part of the global scientific community remains sceptical regarding the use of testosterone replacement therapy (TRT) in men suffering from hypogonadism and prostate cancer (Pca). The negative attitudes with respect to testosterone supplementation in men with hypogonadism and Pca may be justified by the relatively low number of clinical and preclinical studies that specifically dealt with how androgens affect Pca biology. More controversial still is the use of TRT in men in active surveillance or at intermediate or high risk of recurrence and treated by curative radiotherapy. In these clinical scenarios, clinicians should be aware that safety data regarding TRT are scanty limiting our ability to draw definitive conclusions on this important topic. In this review we critically discuss the newest scientific evidence concerning the new challenges in the treatment of men with hypogonadal condition and Pca providing new insights in the pharmacological and psychological approaches.

12.
World J Clin Oncol ; 5(3): 335-47, 2014 Aug 10.
Article in English | MEDLINE | ID: mdl-25114849

ABSTRACT

Metastatic occurrence is the principal cause of death in breast cancer patients. The high osteotropism makes breast cancer the most common primary tumor type associated with metastatic bone disease. The peculiar clinical aspects associated with metastases limited to the skeletal system suggest considering these cases as a distinctive subset of metastatic patients with a better prognosis. Because bone is frequently the first metastatic site in disease relapse, it is feasible that the next improvement in therapeutic options for bone metastatic disease could be associated with an improvement of survival expectation and quality of life in breast cancer patients. Study of the molecular basis of bone remodeling and breast cancer osteotropism has allowed identification of several therapeutic candidates involved in formation and progression of bone metastases. These targets are frequently the determinants of positive feedback between the tumor and bone cells whose clinical outcome is osteolytic lesions. In this review, we discuss the physiopathologic features underlying targeted therapeutic strategies aimed at interfering with the aberrant bone remodeling associated with breast cancer metastases.

13.
Exp Cell Res ; 328(1): 20-31, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128812

ABSTRACT

The protein kinase Src is frequently over-activated in advanced cancers where it modulates the signaling transduction cascade of several growth factors. The feasibility of combination treatment of Src inhibitors with chemotherapy is currently under investigation. We evaluated the anti-tumoral effect of paclitaxel (PTX) in combination with S13, a tyrosine kinase inhibitor with a prevalent specificity for Src, in a hormone-insensible prostate cancer (PCa) cell model. In vivo, combination treatment with PTX and S13 reduced dramatically PCa tumor growth with a relevant difference in the density of new blood vessels with respect to control and single treatments. This reduction was determined by a concomitant impairment of endothelial cell migration and of VEGF release by cancer cells. In fact, S13, when used alone, was sufficient to reduce tubule formation in vivo, and to inhibit VEGFR2 activation and FAK expression in endothelial cells. In addition, the combination treatment determined a significant reduction in ROS production and HIF-1 stabilization in PCa cells respect to single treatments with S13 or PTX. In conclusion, Src-inhibition could be an effective therapeutic strategy aimed at supporting the anti-angiogenic action of PTX in aggressive PCa.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Neovascularization, Pathologic/prevention & control , Paclitaxel/pharmacology , Prostatic Neoplasms/drug therapy , src-Family Kinases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/metabolism , Humans , Immunoenzyme Techniques , Male , Mice , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-2/metabolism , src-Family Kinases/metabolism
14.
BMC Cancer ; 14: 154, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24597899

ABSTRACT

BACKGROUND: Cancer cell adopts peculiar metabolic strategies aimed to sustain the continuous proliferation in an environment characterized by relevant fluctuations in oxygen and nutrient levels. Monocarboxylate transporters MCT1 and MCT4 can drive such adaptation permitting the transport across plasma membrane of different monocarboxylic acids involved in energy metabolism. METHODS: Role of MCTs in tumor-stroma metabolic relationship was investigated in vitro and in vivo using transformed prostate epithelial cells, carcinoma cell lines and normal fibroblasts. Moreover prostate tissues from carcinoma and benign hypertrophy cases were analyzed for individuating clinical-pathological implications of MCT1 and MCT4 expression. RESULTS: Transformed prostate epithelial (TPE) and prostate cancer (PCa) cells express both MCT1 and MCT4 and demonstrated variable dependence on aerobic glycolysis for maintaining their proliferative rate. In glucose-restriction the presence of L-lactate determined, after 24 h of treatment, in PCa cells the up-regulation of MCT1 and of cytochrome c oxidase subunit I (COX1), and reduced the activation of AMP-activated protein kinase respect to untreated cells. The blockade of MCT1 function, performed by si RNA silencing, determined an appreciable antiproliferative effect when L-lactate was utilized as energetic fuel. Accordingly L-lactate released by high glycolytic human diploid fibroblasts WI-38 sustained survival and growth of TPE and PCa cells in low glucose culture medium. In parallel, the treatment with conditioned medium from PCa cells was sufficient to induce glycolytic metabolism in WI-38 cells, with upregulation of HIF-1a and MCT4. Co-injection of PCa cells with high glycolytic WI-38 fibroblasts determined an impressive increase in tumor growth rate in a xenograft model that was abrogated by MCT1 silencing in PCa cells. The possible interplay based on L-lactate shuttle between tumor and stroma was confirmed also in human PCa tissue where we observed a positive correlation between stromal MCT4 and tumor MCT1 expression. CONCLUSIONS: Our data demonstrated that PCa progression may benefit of MCT1 expression in tumor cells and of MCT4 in tumor-associated stromal cells. Therefore, MCTs may result promising therapeutic targets in different phases of neoplastic transformation according to a strategy aimed to contrast the energy metabolic adaptation of PCa cells to stressful environments.


Subject(s)
Lactates/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Stromal Cells/metabolism , Animals , Biological Transport , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Models, Animal , Disease Progression , Fibroblasts/metabolism , Gene Expression , Gene Silencing , Glycolysis , Heterografts , Humans , Male , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/genetics , Symporters/genetics , Symporters/metabolism , Tumor Microenvironment
15.
PLoS One ; 8(11): e79309, 2013.
Article in English | MEDLINE | ID: mdl-24244477

ABSTRACT

The formation of new blood vessels is an essential therapeutic target in many diseases such as cancer, ischemic diseases, and chronic inflammation. In this regard, extremely low-frequency (ELF) electromagnetic fields (EMFs) seem able to inhibit vessel growth when used in a specific window of amplitude. To investigate the mechanism of anti-angiogenic action of ELF-EMFs we tested the effect of a sinusoidal magnetic field (MF) of 2 mT intensity and frequency of 50 Hz on endothelial cell models HUVEC and MS-1 measuring cell status and proliferation, motility and tubule formation ability. MS-1 cells when injected in mice determined a rapid tumor-like growth that was significantly reduced in mice inoculated with MF-exposed cells. In particular, histological analysis of tumors derived from mice inoculated with MF-exposed MS-1 cells indicated a reduction of hemangioma size, of blood-filled spaces, and in hemorrhage. In parallel, in vitro proliferation of MS-1 treated with MF was significantly inhibited. We also found that the MF-exposure down-regulated the process of proliferation, migration and formation of tubule-like structures in HUVECs. Using western blotting and immunofluorescence analysis, we collected data about the possible influence of MF on the signalling pathway activated by the vascular endothelial growth factor (VEGF). In particular, MF exposure significantly reduced the expression and activation levels of VEGFR2, suggesting a direct or indirect influence of MF on VEGF receptors placed on cellular membrane. In conclusion MF reduced, in vitro and in vivo, the ability of endothelial cells to form new vessels, most probably affecting VEGF signal transduction pathway that was less responsive to activation. These findings could not only explain the mechanism of anti-angiogenic action exerted by MFs, but also promote the possible development of new therapeutic applications for treatment of those diseases where excessive angiogenesis is involved.


Subject(s)
Electromagnetic Fields , Magnetic Fields , Neovascularization, Physiologic/radiation effects , Animals , Cell Cycle/radiation effects , Cell Line , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Endothelial Cells/radiation effects , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Neovascularization, Physiologic/physiology , Signal Transduction/radiation effects , Vascular Endothelial Growth Factor Receptor-2/metabolism
17.
PLoS One ; 8(4): e61484, 2013.
Article in English | MEDLINE | ID: mdl-23613861

ABSTRACT

Based on the growing deal of data concerning the biological activity of flavonoid-rich natural products, the aim of the present study was to explore in vitro the potential anti-tumoral activity of Citrus Bergamia (bergamot) juice (BJ), determining its molecular interaction with cancer cells. Here we show that BJ reduced growth rate of different cancer cell lines, with the maximal growth inhibition observed in neuroblastoma cells (SH-SY5Y) after 72 hs of exposure to 5% BJ. The SH-SY5Y antiproliferative effect elicited by BJ was not due to a cytotoxic action and it did not induce apoptosis. Instead, BJ stimulated the arrest in the G1 phase of cell cycle and determined a modification in cellular morphology, causing a marked increase of detached cells. The inhibition of adhesive capacity on different physiologic substrates and on endothelial cells monolayer were correlated with an impairment of actin filaments, a reduction in the expression of the active form of focal adhesion kinase (FAK) that in turn caused inhibition of cell migration. In parallel, BJ seemed to hinder the association between the neural cell adhesion molecule (NCAM) and FAK. Our data suggest a mechanisms through which BJ can inhibit important molecular pathways related to cancer-associated aggressive phenotype and offer new suggestions for further studies on the role of BJ in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Citrus/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Humans
18.
ChemMedChem ; 8(3): 484-96, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23436791

ABSTRACT

Pim-1 is a serine/threonine kinase critically involved in the initiation and progression of various types of cancer, especially leukemia, lymphomas and solid tumors such as prostate, pancreas and colon, and is considered a potential drug target against these malignancies. In an effort to discover new potent Pim-1 inhibitors, a previously identified ATP-competitive indolyl-pyrrolone scaffold was expanded to derive structure-activity relationship data. A virtual screening campaign was also performed, which led to the discovery of additional ATP-competitive inhibitors as well as a series of 2-aminothiazole derivatives, which are noncompetitive with respect to both ATP and peptide substrate. This mechanism of action, which resembles allosteric inhibition, has not previously been characterized for Pim-1. Notably, further evaluation of the 2-aminothiazoles indicated a synergistic inhibitory effect in enzymatic assays when tested in combination with ATP-competitive inhibitors. A synergistic effect in the inhibition of cell proliferation by ATP-competitive and ATP-noncompetitive compounds was also observed in prostate cancer cell lines (PC3), where all Pim-1 inhibitors tested in showed synergism with the known anticancer agent, paclitaxel. These results further establish Pim-1 as a target in cancer therapy, and highlight the potential of these agents for use as adjuvant agents in the treatment of cancer diseases in which Pim-1 is associated with chemotherapeutic resistance.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Antineoplastic Agents/metabolism , Antineoplastic Agents/toxicity , Binding Sites , Binding, Competitive , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Indoles/chemistry , Kinetics , Molecular Docking Simulation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/toxicity , Protein Structure, Tertiary , Proto-Oncogene Proteins c-pim-1/genetics , Proto-Oncogene Proteins c-pim-1/metabolism , Pyrroles/chemistry , Pyrroles/metabolism , Pyrroles/toxicity , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/metabolism , Thiazoles/toxicity
19.
ACS Med Chem Lett ; 4(7): 622-6, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-24900720

ABSTRACT

Design and synthesis of prodrugs of promising drug candidates represents a valid strategy to overcome the lack of favorable ADME properties, in particular aqueous solubility and bioavailability. We report herein the successful application of this strategy with two representative pyrazolo[3,4-d]pyrimidine derivatives (1 and 2), which led to the development of the corresponding and highly water-soluble antitumor prodrugs (7 and 8). In vitro studies confirmed a significant improvement of aqueous solubility and, for compound 8, good plasma stability, suggesting superior in vivo bioavailability. As expected, the uncleaved water-soluble prodrugs 7 and 8 showed no activity toward the enzymatic targets (c-Src and c-Abl) but revealed promising antiproliferative activity in myeloid cell lines, as a consequence of the in vitro hydrolysis of the selected solubilizing moiety, followed by the release of the active compounds (1 and 2).

20.
Diagn Pathol ; 6: 34, 2011 Apr 13.
Article in English | MEDLINE | ID: mdl-21489246

ABSTRACT

BACKGROUND: Nowadays, the histological examination of prostate core needle biopsies is still regarded as the gold standard in the diagnosis of prostate cancer (PCa). We investigated if the tissue print of core needle biopsy (biopsy print) could be used as adjunctive molecular investigative procedures in conjunction with routine histological examination of biopsy to improve PCa diagnosis. METHODS: The direct contact of PCa core biopsy to nitrocellulose membrane resulted in the release of a cellular micropeel that was used for downstream analytical procedures. RESULTS: By zymogram print-phoresis we demonstrated that matrix metalloproteases MMP-2 and MMP-9 could be visualized in biopsy prints and that the gelatinolytic activity was positively correlated with immunohistochemistry analysis of the same markers in matched bioptic specimens. Moreover, we compared the ability to detect the PCa-associated hypermethylation of GSTP1 promoter in DNA extracted from biopsy prints with those of the corresponding core needle biopsies. Biopsy prints demonstrated the same specificity of biopsies in detecting PCa (50%) while the sensitivity and the positive predictive value were lower than biopsies (56% vs 78% and 63% vs 70%, respectively). CONCLUSIONS: Biopsy print, combining a molecular point of view to the routinely hystopathological analysis of prostate biopsies, should be a useful tool to improve the diagnosis of PCa.


Subject(s)
Biopsy, Needle/methods , Diagnostic Techniques and Procedures , Prostate/pathology , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Aged , Biomarkers, Tumor/metabolism , Collodion , Humans , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Prostatic Neoplasms/metabolism , Retrospective Studies , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...