52 research outputs found

    Heme oxygenase-1 in central nervous system malignancies

    Get PDF
    Central nervous system tumors are the most common pediatric solid tumors and account for 20%-25% of all childhood malignancies. Several lines of evidence suggest that brain tumors show altered redox homeostasis that triggers the activation of various survival pathways, leading to disease progression and chemoresistance. Among these pathways, heme oxygenase-1 (HO-1) plays an important role. HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. The biological effects of HO-1 in tumor cells have been shown to be cell-specific since, in some tumors, its upregulation promotes cell cycle arrest and cellular death, whereas, in other neoplasms, it is associated with tumor survival and progression. This review focuses on the role of HO-1 in central nervous system malignancies and the possibility of exploiting such a target to improve the outcome of well-established therapeutic regimens. Finally, several studies show that HO-1 overexpression is involved in the development and resistance of brain tumors to chemotherapy and radiotherapy, suggesting the use of HO-1 as an innovative therapeutic target to overcome drug resistance. The following keywords were used to search the literature related to this topic: nuclear factor erythroid 2 p45-related factor 2, heme oxygenase, neuroblastoma, medulloblastoma, meningioma, astrocytoma, oligodendroglioma, glioblastoma multiforme, and gliomas

    Radicalization in Correctional Systems: A Scoping Review of the Literature Evaluating the Effectiveness of Preventing and Countering Interventions

    Get PDF
    Although several prevention and countering radicalization programs in correctional institutions have been carried out in Europe and worldwide, little is known about their effectiveness. Thus, the current scoping review aimed at reporting on the state of the art of the literature assessing and evaluating such actions, identifying knowledge gaps, and examining methods used to assess their successfulness and impact. A total of eight studies that met eligibility criteria were reviewed after performing a search on Scopus, Web of Science, and PsychInfo (Ebsco) databases: Two studies evaluated a reintegration initiative based in the Netherlands; four focused on an Australian disengagement program; and two offered insights on a rehabilitation program carried out in Sri Lanka. Findings were discussed according to their evaluation methods (qualitative, mixed-methods, and quantitative) and instruments. Results highlighted that the evaluation process is still confronted with several methodological difficulties, such as a lack of agreement on how to univocally define and measure success and the identification of uniform indicators of deradicalization and disengagement. Additionally, results revealed that the effectiveness of these interventions is predominantly based on anecdotal evidence rather than on rigorous, empirical proofs. As such, it was not possible to compare these programs and determine which worked best. Suggestions for future research and practical implications for policymakers, prison governors, and practitioners are offered in the concluding section of this work.   Acknowledgements The present study was made possible by funding from SERENY (“Strengthening approaches for the prevention of youth radicalisation in prison and probation settings”), a project co-funded by the Justice Program of the European Union, GA no. 101007425. The contents of this publication are the sole responsibility of the authors and can in no way be taken to reflect the views of the European Commission

    Microfluidics as a Novel Tool for Biological and Toxicological Assays in Drug Discovery Processes: Focus on Microchip Electrophoresis

    Get PDF
    This work is licensed under a Creative Commons Attribution 4.0 International License.The last decades of biological, toxicological, and pharmacological research have deeply changed the way researchers select the most appropriate ‘pre-clinical model’. The absence of relevant animal models for many human diseases, as well as the inaccurate prognosis coming from ‘conventional’ pre-clinical models, are among the major reasons of the failures observed in clinical trials. This evidence has pushed several research groups to move more often from a classic cellular or animal modeling approach to an alternative and broader vision that includes the involvement of microfluidic-based technologies. The use of microfluidic devices offers several benefits including fast analysis times, high sensitivity and reproducibility, the ability to quantitate multiple chemical species, and the simulation of cellular response mimicking the closest human in vivo milieu. Therefore, they represent a useful way to study drug–organ interactions and related safety and toxicity, and to model organ development and various pathologies ‘in a dish’. The present review will address the applicability of microfluidic-based technologies in different systems (2D and 3D). We will focus our attention on applications of microchip electrophoresis (ME) to biological and toxicological studies as well as in drug discovery and development processes. These include high-throughput single-cell gene expression profiling, simultaneous determination of antioxidants and reactive oxygen and nitrogen species, DNA analysis, and sensitive determination of neurotransmitters in biological fluids. We will discuss new data obtained by ME coupled to laser-induced fluorescence (ME-LIF) and electrochemical detection (ME-EC) regarding the production and degradation of nitric oxide, a fundamental signaling molecule regulating virtually every critical cellular function. Finally, the integration of microfluidics with recent innovative technologies—such as organoids, organ-on-chip, and 3D printing—for the design of new in vitro experimental devices will be presented with a specific attention to drug development applications. This ‘composite’ review highlights the potential impact of 2D and 3D microfluidic systems as a fast, inexpensive, and highly sensitive tool for high-throughput drug screening and preclinical toxicological studies.Italian Ministry of Health Research Program 2018 (2635256)American Heart Association-Midwest Affiliate Postdoctoral Research Fellowship (NFP0075515)Italian Ministry of Economic Development (F/200110/02/X45)Italian Ministry of EducationNIH COBRE P20GM103638Oasi Research Institute—IRCC

    Anti-proliferative effects of Cetuximab and Trastuzumab in colorectal cancer cell lines

    Get PDF
    Colon cancer is one of the most common human malignancies and a leading cause of death worldwide. In Europe around 250.000 new colon cases are diagnosed each year, accounting for around 9% of all the malignancies (Labianca R et al., 2010; Berrino F et al., 2007). Dysregulation of the signalling pathways induced through EGF receptors (ErbB/ HER receptors) by their over-expression or constitutive activation can promote tumor processes (Lurje G et al., 2009), including colorectal cancer. Therefore, the ErbB/HER receptor family with their most prominent members EGFR and HER-2 represents validated targets for anti-cancer therapy. Cetuximab and trastuzumab are two monoclonal antibodies approved for treating, respectively, metastatic colorectal and breast cancer. Because the monotherapy with cetuximab in metastatic colorectal is often insufficient (Cunningham D et al., 2004), it is useful to develop complementary therapeutic strategies to enhance antibody efficacy. A possible approach is co-administration of inhibitors, targeting multiple members of the EGF receptor family. In this study we examined the effect of cetuximab and trastuzumab in combination using two human colon cancer cell lines as a model. We observed that the two drugs had a cytostatic effect and inhibited the proliferation of both the cell lines in a time- and concentration-dependent manner. However, the combination had lower efficacy on one cell line than the other, with growth inhibition of 31% in the former and 49% in the latter. This result was associated to specific changes in cell cycle distribution, while no apoptosis was observed. Chromosome copy number heterogeneity and aneuploidy in tumoral cell lines have been reported (Pellestor F et al., 1999). Our data deriving from the cell cycle analysis confirmed the aneuploidy and polyploidy in our cellular models and are useful to explain cellular response to the combination. We used fluorescent in situ hybridisation analysis to evaluate EGFR and HER-2 gene amplification status. Both the tumour cell lines resulted in an abnormal copy number for the two genes resulting from aneuploidy (polisomy of chromosome 7 and 17) which is not responsible for the difference in sensitivity to cetuximab and trastuzumab between the two cell lines. In order to understand and to improve the pharmacological efficacy of cetuximab and trastuzumab combination, it will be useful to elucidate the molecular mechanisms involved in their activity. This will allow to develop novel and interesting approaches to cancer therapy

    Recent advances in molecular diagnostics of colorectal cancer by genomic arrays: proposal for a procedural shift in biological sampling and pathological report

    Get PDF
    Two forms of genetic instability have been described in colorectal cancer: chromosomal instability, characterized by structural and numerical chromosomal abnormalities and associated to aneuploidy; and microsatellite instability, characterized by a deficiency in the mismatch repair system that leads to slippage in microsatellites and is associated to euploidy. Thirteen colorectal cancer sample DNAs were analyzed after colectomy. High-resolution genome-wide DNA copy number and Single Nucleotide Polimorphism genotyping analysis was performed by Affymetrix SNP 6.0 arrays that interrogates 906,600 single nucleotide polymorphisms and 945,826 copy number probes. We implemented this analysis as part of a routine procedure that includes the sampling of fresh tissue from the tumor mass without affecting the subsequent standard histopathological procedure. The novel molecular technology allows the determination of a genome-wide molecular karyotype using only 500 ng of high-quality tumor DNA; it distinguishes the two main types of genomic instability, discriminating between chromosomal instability positive and negative tumors. It also detects loss of heterozygosity (LOH) regions, called copy neutral-LOH. Tumor-associated copy neutral-LOH regions may play a pivotal role in oncogenesis when they determine duplications of either activating or loss of function gene mutation. We observed recurrent gains of chromosomes 2, 7, 8q, 9, 12, 13, 20 and losses of chromosomes 4, 5, 8p, 15, 17p, 18, 22, and Y, in agreement with previous cytogenetic studies. The use of such sampling procedure could stimulate the routine detection of point mutations in specific genes, thus avoiding subsequent sectioning of formalin-fixed and paraffin-embedded samples

    Modulation of Pro-Oxidant and Pro-Inflammatory Activities of M1 Macrophages by the Natural Dipeptide Carnosine

    Get PDF
    This work is licensed under a Creative Commons Attribution 4.0 International License.Carnosine is a natural endogenous dipeptide widely distributed in mammalian tissues, existing at particularly high concentrations in the muscles and brain and possesses well-characterized antioxidant and anti-inflammatory activities. In an in vitro model of macrophage activation, induced by lipopolysaccharide + interferon-gamma (LPS + IFN-γ), we here report the ability of carnosine to modulate pro-oxidant and pro-inflammatory activities of macrophages, representing the primary cell type that is activated as a part of the immune response. An ample set of parameters aimed to evaluate cytotoxicity (MTT assay), energy metabolism (HPLC), gene expressions (high-throughput real-time PCR (qRT-PCR)), protein expressions (western blot) and nitric oxide production (qRT-PCR and HPLC), was used to assess the effects of carnosine on activated macrophages challenged with a non cytotoxic LPS (100 ng/mL) + IFN-γ (600 U/mL) concentration. In our experimental model, main carnosine beneficial effects were: (1) the modulation of nitric oxide production and metabolism; (2) the amelioration of the macrophage energy state; (3) the decrease of the expressions of pro-oxidant enzymes (Nox-2, Cox-2) and of the lipid peroxidation product malondialdehyde; (4) the restoration and/or increase of the expressions of antioxidant enzymes (Gpx1, SOD-2 and Cat); (5) the increase of the transforming growth factor-β1 (TGF-β1) and the down-regulation of the expressions of interleukins 1β and 6 (IL-1β and IL-6) and 6) the increase of the expressions of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1). According to these results carnosine is worth being tested in the treatment of diseases characterized by elevated levels of oxidative stress and inflammation (atherosclerosis, cancer, depression, metabolic syndrome, and neurodegenerative diseases)

    Carnosine Protects Macrophages against the Toxicity of Aβ1-42 Oligomers by Decreasing Oxidative Stress

    Get PDF
    Carnosine (β-alanyl-L-histidine) is a naturally occurring endogenous peptide widely distributed in excitable tissues such as the brain. This dipeptide has well-known antioxidant, anti-inflammatory, and anti-aggregation activities, and it may be useful for treatment of neurodegenerative disorders such as Alzheimer’s disease (AD). In this disease, peripheral infiltrating macrophages play a substantial role in the clearance of amyloid beta (Aβ) peptides from the brain. Correspondingly, in patients suffering from AD, defects in the capacity of peripheral macrophages to engulf Aβ have been reported. The effects of carnosine on macrophages and oxidative stress associated with AD are consequently of substantial interest for drug discovery in this field. In the present work, a model of stress induced by Aβ1-42 oligomers was investigated using a combination of methods including trypan blue exclusion, microchip electrophoresis with laser-induced fluorescence, flow cytometry, fluorescence microscopy, and high-throughput quantitative real-time PCR. These assays were used to assess the ability of carnosine to protect macrophage cells, modulate oxidative stress, and profile the expression of genes related to inflammation and pro- and antioxidant systems. We found that pre-treatment of RAW 264.7 macrophages with carnosine counteracted cell death and apoptosis induced by Aβ1-42 oligomers by decreasing oxidative stress as measured by levels of intracellular nitric oxide (NO)/reactive oxygen species (ROS) and production of peroxynitrite. This protective activity of carnosine was not mediated by modulation of the canonical inflammatory pathway but instead can be explained by the well-known antioxidant and free-radical scavenging activities of carnosine, enhanced macrophage phagocytic activity, and the rescue of fractalkine receptor CX3CR1. These new findings obtained with macrophages challenged with Aβ1-42 oligomers, along with the well-known multimodal mechanism of action of carnosine in vitro and in vivo, substantiate the therapeutic potential of this dipeptide in the context of AD pathology

    Carnosine Decreases PMA-Induced Oxidative Stress and Inflammation in Murine Macrophages

    Get PDF
    This work is licensed under a Creative Commons Attribution 4.0 International License.Carnosine is an endogenous dipeptide composed of β-alanine and L-histidine. This naturally occurring molecule is present at high concentrations in several mammalian excitable tissues such as muscles and brain, while it can be found at low concentrations in a few invertebrates. Carnosine has been shown to be involved in different cellular defense mechanisms including the inhibition of protein cross-linking, reactive oxygen and nitrogen species detoxification as well as the counteraction of inflammation. As a part of the immune response, macrophages are the primary cell type that is activated. These cells play a crucial role in many diseases associated with oxidative stress and inflammation, including atherosclerosis, diabetes, and neurodegenerative diseases. In the present study, carnosine was first tested for its ability to counteract oxidative stress. In our experimental model, represented by RAW 264.7 macrophages challenged with phorbol 12-myristate 13-acetate (PMA) and superoxide dismutase (SOD) inhibitors, carnosine was able to decrease the intracellular concentration of superoxide anions (O2−•) as well as the expression of Nox1 and Nox2 enzyme genes. This carnosine antioxidant activity was accompanied by the attenuation of the PMA-induced Akt phosphorylation, the down-regulation of TNF-α and IL-6 mRNAs, and the up-regulation of the expression of the anti-inflammatory mediators IL-4, IL-10, and TGF-β1. Additionally, when carnosine was used at the highest dose (20 mM), there was a generalized amelioration of the macrophage energy state, evaluated through the increase both in the total nucleoside triphosphate concentrations and the sum of the pool of intracellular nicotinic coenzymes. Finally, carnosine was able to decrease the oxidized (NADP+)/reduced (NADPH) ratio of nicotinamide adenine dinucleotide phosphate in a concentration dependent manner, indicating a strong inhibitory effect of this molecule towards the main source of reactive oxygen species in macrophages. Our data suggest a multimodal mechanism of action of carnosine underlying its beneficial effects on macrophage cells under oxidative stress and inflammation conditions

    PARP-14 Promotes Survival of Mammalian ι but Not β Pancreatic Cells Following Cytokine Treatment

    Get PDF
    PARP-14 (poly-ADP Ribose Polymerase-14), a member of the PARP family, belongs to the group of Bal proteins (B Aggressive Lymphoma). PARP-14 has recently appeared to be involved in the transduction pathway mediated by JNKs (c Jun N terminal Kinases), among which JNK2 promotes cancer cell survival. Several pharmacological PARP inhibitors are currently used as antitumor agents, even though they have also proved to be effective in many inflammatory diseases. Cytokine release from immune system cells characterizes many autoimmune inflammatory disorders, including type I diabetes, in which the inflammatory state causes β cell loss. Nevertheless, growing evidence supports a concomitant implication of glucagon secreting ι cells in type I diabetes progression. Here, we provide evidence on the activation of a survival pathway, mediated by PARP-14, in pancreatic ι cells, following treatment of ιTC1.6 glucagonoma and βTC1 insulinoma cell lines with a cytokine cocktail: interleukin 1 beta (IL-1β), interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-ι). Through qPCR, western blot and confocal analysis, we demonstrated higher expression levels of PARP-14 in ιTC1.6 cells with respect to βTC1 cells under inflammatory stimuli. By cytofluorimetric and caspase-3 assays, we showed the higher resistance of ι cells compared to β cells to apoptosis induced by cytokines. Furthermore, the ability of PJ-34 to modulate the expression of the proteins involved in the survival pathway suggests a protective role of PARP-14. These data shed light on a poorly characterized function of PARP-14 in ιTC1.6 cells in inflammatory contexts, widening the potential pharmacological applications of PARP inhibitors
    • …
    corecore