Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(14)2022 Jul 16.
Article in English | MEDLINE | ID: mdl-35884524

ABSTRACT

Recent reports have shown a link between radiation exposure and non-cancer diseases such as radiation-induced heart disease (RIHD). Radiation exposures are often inhomogeneous, and out-of-target effects have been studied in terms of cancer risk, but very few studies have been carried out for non-cancer diseases. Here, the role of miRNAs in the pathogenesis of RIHD was investigated. C57Bl/6J female mice were whole- (WBI) or partial-body-irradiated (PBI) with 2 Gy of X-rays or sham-irradiated (SI). In PBI exposure, the lower third of the mouse body was irradiated, while the upper two-thirds were shielded. From all groups, hearts were collected 15 days or 6 months post-irradiation. The MiRNome analysis at 15 days post-irradiation showed that miRNAs, belonging to the myomiR family, were highly differentially expressed in WBI and PBI mouse hearts compared with SI hearts. Raman spectral data collected 15 days and 6 months post-irradiation showed biochemical differences among SI, WBI and PBI mouse hearts. Fibrosis in WBI and PBI mouse hearts, indicated by the increased deposition of collagen and the overexpression of genes involved in myofibroblast activation, was found 6 months post-irradiation. Using an in vitro co-culture system, involving directly irradiated skeletal muscle and unirradiated ventricular cardiac human cells, we propose the role of miR-1/133a as mediators of the abscopal response, suggesting that miRNA-based strategies could be relevant for limiting tissue-dependent reactions in non-directly irradiated tissues.

2.
Syst Rev ; 11(1): 29, 2022 02 16.
Article in English | MEDLINE | ID: mdl-35172887

ABSTRACT

BACKGROUND: An Italian project aims to review the scientific literature on the possible carcinogenicity of radiofrequency (100 kHz-300 GHz) electromagnetic field (RF-EMF) exposure. The ENEA team has to carry out a systematic review of the in vivo studies on this topic. OBJECTIVES: Development of a protocol for a systematic review (meta-analysis included) to investigate the potential carcinogenic risk following RF-EMF in vivo exposure to doses above or within legal limits. The aims of this review are (1) to provide a descriptive and, if possible, a quantitative summary of the results of the examined RF-EMF in vivo studies, together with an assessment of the consistency of observations and of the causes of heterogeneity, and (2) to assess the weight of evidence to support or refute the hypothesis of carcinogenic effects caused by RF-EMF exposure and to draw conclusions about the potential for carcinogenicity of RF-EMF exposure. METHODS: We will search for relevant studies in electronic academic databases and in the reference list of selected papers and reviews on the topic, including the descriptive reviews on RF-EMF carcinogenic effect carried out by international panels of experts since 2011. The following elements of the PECO question were defined: experimental studies on rodents of both sexes, all ages and species, all genetic backgrounds (Population) exposed to RF-EMF alone, or in combination with other physical or chemical agents (Exposure); only studies reporting outcome data in exposed and sham control groups (Comparison); and all types of cancer with all tumor-related outcome measures (Outcome) will be included. Only peer-reviewed articles written in English will be considered without limit in the publication date. Eligibility criteria were defined for papers to be included. A risk of bias assessment will be performed using a tool specifically developed for animal studies. A meta-analysis will be performed, if feasible, for all outcome measures; for subgroup analysis, a minimum of 3 studies per subgroup will be required. If meta-analysis will not be possible, a narrative synthesis of the results will be reported. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD42020191105 HIGHLIGHTS: An Italian collaborative research agreement aims to review the scientific literature on the possible carcinogenicity of RF-EMF (100 kHz - 300 GHz). The ENEA team will systematically review and, if possible, meta-analyse estimates the effects of in vivo exposure to RF-EMF exposure on cancer. The ENEA group is a multidisciplinary team of researchers with a consolidated experience both in carcinogenicity experiments and radiofrequency dosimetric assessment. The proposed protocol uses the NTP OHAT Approach for Systematic Review as an organizing framework. The proposed protocol aims to lead to the first systematic review providing a strength of evidence assessment on this topic.


Subject(s)
Electromagnetic Fields , Neoplasms , Animals , Electromagnetic Fields/adverse effects , Female , Male , Meta-Analysis as Topic , Outcome Assessment, Health Care , Radio Waves/adverse effects , Research Design , Systematic Reviews as Topic
3.
Int J Mol Sci ; 23(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35216284

ABSTRACT

Cell communication via exosomes is capable of influencing cell fate in stress situations such as exposure to ionizing radiation. In vitro and in vivo studies have shown that exosomes might play a role in out-of-target radiation effects by carrying molecular signaling mediators of radiation damage, as well as opposite protective functions resulting in resistance to radiotherapy. However, a global understanding of exosomes and their radiation-induced regulation, especially within the context of an intact mammalian organism, has been lacking. In this in vivo study, we demonstrate that, compared to sham-irradiated (SI) mice, a distinct pattern of proteins and miRNAs is found packaged into circulating plasma exosomes after whole-body and partial-body irradiation (WBI and PBI) with 2 Gy X-rays. A high number of deregulated proteins (59% of WBI and 67% of PBI) was found in the exosomes of irradiated mice. In total, 57 and 13 miRNAs were deregulated in WBI and PBI groups, respectively, suggesting that the miRNA cargo is influenced by the tissue volume exposed to radiation. In addition, five miRNAs (miR-99b-3p, miR-200a-3p, miR-200a, miR-182-5p, miR-182) were commonly overexpressed in the exosomes from the WBI and PBI groups. In this study, particular emphasis was also given to the determination of the in vivo effect of exosome transfer by intracranial injection in the highly radiosensitive neonatal cerebellum at postnatal day 3. In accordance with a major overall anti-apoptotic function of the commonly deregulated miRNAs, here, we report that exosomes from the plasma of irradiated mice, especially in the case of WBI, prevent radiation-induced apoptosis, thus holding promise for exosome-based future therapeutic applications against radiation injury.


Subject(s)
Exosomes , MicroRNAs , Radiation Injuries , Animals , Apoptosis , Cerebellum/metabolism , Exosomes/metabolism , Mammals/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Proteomics , Radiation Injuries/metabolism
4.
Int J Mol Sci ; 22(19)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34638864

ABSTRACT

Medulloblastoma (MB) is a primary central nervous system tumor affecting mainly young children. New strategies of drug delivery are urgent to treat MB and, in particular, the SHH-dependent subtype-the most common in infants-in whom radiotherapy is precluded due to the severe neurological side effects. Plant virus nanoparticles (NPs) represent an innovative solution for this challenge. Tomato bushy stunt virus (TBSV) was functionally characterized as a carrier for drug targeted delivery to a murine model of Shh-MB. The TBSV NPs surface was genetically engineered with peptides for brain cancer cell targeting, and the modified particles were produced on a large scale using Nicotiana benthamiana plants. Tests on primary cultures of Shh-MB cells allowed us to define the most efficient peptides able to induce specific uptake of TBSV. Immunofluorescence and molecular dynamics simulations supported the hypothesis that the specific targeting of the NPs was mediated by the interaction of the peptides with their natural partners and reinforced by the presentation in association with the virus. In vitro experiments demonstrated that the delivery of Doxorubicin through the chimeric TBSV allowed reducing the dose of the chemotherapeutic agent necessary to induce a significant decrease in tumor cells viability. Moreover, the systemic administration of TBSV NPs in MB symptomatic mice, independently of sex, confirmed the ability of the virus to reach the tumor in a specific manner. A significant advantage in the recognition of the target appeared when TBSV NPs were functionalized with the CooP peptide. Overall, these results open new perspectives for the use of TBSV as a vehicle for the targeted delivery of chemotherapeutics to MB in order to reduce early and late toxicity.


Subject(s)
Cerebellar Neoplasms , Doxorubicin , Drug Delivery Systems , Hedgehog Proteins/metabolism , Medulloblastoma , Nanoparticles , Neoplasm Proteins/metabolism , Tombusvirus/chemistry , Animals , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Hedgehog Proteins/genetics , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Mutant Strains , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Neoplasm Proteins/genetics , Nicotiana/virology
5.
Int J Mol Sci ; 22(8)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33924260

ABSTRACT

The brain undergoes ionizing radiation exposure in many clinical situations, particularly during radiotherapy for brain tumors. The critical role of the hippocampus in the pathogenesis of radiation-induced neurocognitive dysfunction is well recognized. The goal of this study is to test the potential contribution of non-targeted effects in the detrimental response of the hippocampus to irradiation and to elucidate the mechanisms involved. C57Bl/6 mice were whole body (WBI) or partial body (PBI) irradiated with 0.1 or 2.0 Gy of X-rays or sham irradiated. PBI consisted of the exposure of the lower third of the mouse body, whilst the upper two thirds were shielded. Hippocampi were collected 15 days or 6 months post-irradiation and a multi-omics approach was adopted to assess the molecular changes in non-coding RNAs, proteins and metabolic levels, as well as histological changes in the rate of hippocampal neurogenesis. Notably, at 2.0 Gy the pattern of early molecular and histopathological changes induced in the hippocampus at 15 days following PBI were similar in quality and quantity to the effects induced by WBI, thus providing a proof of principle of the existence of out-of-target radiation response in the hippocampus of conventional mice. We detected major alterations in DAG/IP3 and TGF-ß signaling pathways as well as in the expression of proteins involved in the regulation of long-term neuronal synaptic plasticity and synapse organization, coupled with defects in neural stem cells self-renewal in the hippocampal dentate gyrus. However, compared to the persistence of the WBI effects, most of the PBI effects were only transient and tended to decrease at 6 months post-irradiation, indicating important mechanistic difference. On the contrary, at low dose we identified a progressive accumulation of molecular defects that tended to manifest at later post-irradiation times. These data, indicating that both targeted and non-targeted radiation effects might contribute to the pathogenesis of hippocampal radiation-damage, have general implications for human health.


Subject(s)
Cranial Irradiation , Hippocampus/metabolism , Hippocampus/radiation effects , Metabolome , Neurogenesis/genetics , Neurogenesis/radiation effects , Proteome , Transcriptome , Animals , Computational Biology/methods , Cranial Irradiation/adverse effects , Female , Gene Expression Regulation , Immunohistochemistry , Mice , Radiation Dosage , Signal Transduction
6.
Int J Radiat Oncol Biol Phys ; 109(5): 1495-1507, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33509660

ABSTRACT

PURPOSE: Cancer stem cells constitute an endless reserve for the maintenance and progression of tumors, and they could be the reason for conventional therapy failure. New therapeutic strategies are necessary to specifically target them. In this context, microsecond pulsed electric fields have been selected to expose D283Med cells, a human medulloblastoma cell line resulted to be rich in cancer stem cells, and normal human astrocytes. METHODS: We analyzed in vitro different endpoints at different times after microsecond pulsed electric field exposure, such as permeabilization, reactive oxygen species generation, cell viability/proliferation, cell cycle, and clonogenicity, as well as the expression of different genes involved in cell cycle, apoptosis, and senescence. Furthermore, the response of D283Med cells exposed to microsecond pulsed electric fields was validated in vivo in a heterotopic mouse xenograft model. RESULTS: Our in vitro results showed that a specific pulse protocol (ie, 0.3 MV/m, 40 µs, 5 pulses) was able to induce irreversible membrane permeabilization and apoptosis exclusively in medulloblastoma cancer stem cells. In the surviving cells, reactive oxygen species generation was observed, together with a transitory G2/M cell-cycle arrest with a senescence-associated phenotype via the upregulation of GADD45A. In vivo results, after pulsed electric field exposure, demonstrated a significant tumor volume reduction with no eradication of tumor mass. In conjunction, we verified the efficacy of electric pulse pre-exposure followed by ionizing irradiation in vivo to enable complete inhibition of tumor growth. CONCLUSIONS: Our data reveal novel therapeutic options for the targeting of medulloblastoma cancer stem cells, indicating nonionizing pulsed electric field pre-exposure as an effective means to overcome the radioresistance of cancer stem cells.


Subject(s)
Cerebellar Neoplasms/therapy , Electroporation/methods , Medulloblastoma/therapy , Neoplastic Stem Cells/physiology , Animals , Apoptosis/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation , Cell Survival , Cellular Senescence/genetics , Cerebellar Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints/genetics , Genes, cdc , Humans , M Phase Cell Cycle Checkpoints/genetics , Medulloblastoma/pathology , Mice , Mice, Nude , Radiation Tolerance , Reactive Oxygen Species/metabolism , Tumor Burden , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
7.
Int J Mol Sci ; 21(21)2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33182277

ABSTRACT

Molecular communication between irradiated and unirradiated neighbouring cells initiates radiation-induced bystander effects (RIBE) and out-of-field (abscopal) effects which are both an example of the non-targeted effects (NTE) of ionising radiation (IR). Exosomes are small membrane vesicles of endosomal origin and newly identified mediators of NTE. Although exosome-mediated changes are well documented in radiation therapy and oncology, there is a lack of knowledge regarding the role of exosomes derived from inside and outside the radiation field in the early and delayed induction of NTE following IR. Therefore, here we investigated the changes in exosome profile and the role of exosomes as possible molecular signalling mediators of radiation damage. Exosomes derived from organs of whole body irradiated (WBI) or partial body irradiated (PBI) mice after 24 h and 15 days post-irradiation were transferred to recipient mouse embryonic fibroblast (MEF) cells and changes in cellular viability, DNA damage and calcium, reactive oxygen species and nitric oxide signalling were evaluated compared to that of MEF cells treated with exosomes derived from unirradiated mice. Taken together, our results show that whole and partial-body irradiation increases the number of exosomes, instigating changes in exosome-treated MEF cells, depending on the source organ and time after exposure.


Subject(s)
Exosomes/radiation effects , Radiation Injuries/pathology , Animals , Bystander Effect/radiation effects , Calcium/metabolism , Cell Survival/radiation effects , Cells, Cultured , DNA Damage/radiation effects , Exosomes/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/radiation effects , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Radiation Injuries/metabolism , Radiation, Ionizing , Reactive Oxygen Species/metabolism , Signal Transduction/radiation effects
8.
Cancers (Basel) ; 12(1)2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31963405

ABSTRACT

Medulloblastoma (MB) is the most common malignant pediatric brain tumor. Despite the progress of new treatments, the risk of recurrence, morbidity, and death remains significant and the long-term adverse effects in survivors are substantial. The fraction of cancer stem-like cells (CSCs) because of their self-renewal ability and multi-lineage differentiation potential is critical for tumor initiation, growth, and resistance to therapies. For the development of new CSC-targeted therapies, further in-depth studies are needed using enriched and stable MB-CSCs populations. This work, aimed at identifying the amount of CSCs in three available human cell lines (DAOY, D341, and D283), describes different approaches based on the expression of stemness markers. First, we explored potential differences in gene and protein expression patterns of specific stem cell markers. Then, in order to identify and discriminate undifferentiated from differentiated cells, MB cells were characterized using a physical characterization method based on a high-frequency dielectrophoresis approach. Finally, we compared their tumorigenic potential in vivo, through engrafting in nude mice. Concordantly, our findings identified the D283 human cell line as an ideal model of CSCs, providing important evidence on the use of a commercial human MB cell line for the development of new strategic CSC-targeting therapies.

10.
Oncotarget ; 8(60): 100958-100974, 2017 Nov 24.
Article in English | MEDLINE | ID: mdl-29254138

ABSTRACT

Mutations in DNA repair pathways are frequent in human cancers. Hence, gaining insights into the interaction of DNA repair genes is key to development of novel tumor-specific treatment strategies. In this study, we tested the functional relationship in development and oncogenesis between the homologous recombination (HR) factor Rad54 and Parp-1, a nuclear enzyme that plays a multifunctional role in DNA damage signaling and repair. We introduced single or combined Rad54 and Parp-1 inactivating germline mutations in Ptc1 heterozygous mice, a well-characterized model of medulloblastoma, the most common malignant pediatric brain tumor. Our study reveals that combined inactivation of Rad54 and Parp-1 causes a marked growth delay culminating in perinatallethality, providing for the first time evidence of synthetic lethal interactions between Rad54 and Parp-1 in vivo. Although the double mutation hampered investigation of Rad54 and Parp-1 interactions in cerebellum tumorigenesis, insights were gained by showing accumulation of endogenous DNA damage and increased apoptotic rate in granule cell precursors (GCPs). A network-based approach to detect differential expression of DNA repair genes in the cerebellum revealed perturbation of p53 signaling in Rad54-/-/Parp-1-/-/Ptc1+/-, and MEFs from combined Rad54/Parp-1 mutants showed p53/p21-dependent typical senescent features. These findings help elucidate the genetic interplay between Rad54 and Parp-1 by suggesting that p53/p21-mediated apoptosis and/or senescence may be involved in synthetic lethal interactions occurring during development and inhibition of tumor growth.

11.
Sci Rep ; 7(1): 14238, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079783

ABSTRACT

Medulloblastoma (MB) is the most common pediatric brain tumor, comprising four distinct molecular variants, one of which characterized by activation of the Sonic Hedgehog (SHH) pathway, driving 25-30% of sporadic MB. SHH-dependent MBs arise from granule cell precursors (GCPs), are fatal in 40-70% of cases and radioresistance strongly contributes to poor prognosis and tumor recurrence. Patched1 heterozygous (Ptch1 +/-) mice, carrying a germ-line heterozygous inactivating mutation in the Ptch1 gene, the Shh receptor and negative regulator of the pathway, are uniquely susceptible to MB development after radiation damage in neonatal cerebellum. Here, we irradiated ex-vivo GCPs isolated from cerebella of neonatal WT and Ptch1 +/- mice. Our results highlight a less differentiated status of Ptch1-mutated cells after irradiation, influencing DNA damage response. Increased expression levels of pluripotency genes Nanog, Oct4 and Sal4, together with greater clonogenic potential, clearly suggest that radiation induces expansion of the stem-like cell compartment through cell-reprogramming and self-renewal maintenance, and that this mechanism is strongly dependent on Nanog. These results contribute to clarify the molecular mechanisms that control radiation-induced Shh-mediated tumorigenesis and may suggest Nanog as a potential target to inhibit for adjuvant radiotherapy in treatment of SHH-dependent MB.


Subject(s)
Cell Self Renewal/radiation effects , Cellular Reprogramming/radiation effects , Medulloblastoma/pathology , Nanog Homeobox Protein/metabolism , Patched-1 Receptor/deficiency , Patched-1 Receptor/metabolism , Animals , Apoptosis/radiation effects , Carcinogenesis/radiation effects , Cell Differentiation/radiation effects , Cell Line, Tumor , DNA Damage , Dose-Response Relationship, Radiation , Gene Knockout Techniques , Mice , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Patched-1 Receptor/genetics
12.
Oncotarget ; 7(42): 68253-68269, 2016 Oct 18.
Article in English | MEDLINE | ID: mdl-27626168

ABSTRACT

It has historically been accepted that incorrectly repaired DNA double strand breaks (DSBs) are the principal lesions of importance regarding mutagenesis, and long-term biological effects associated with ionizing radiation. However, radiation may also cause dysregulation of epigenetic processes that can lead to altered gene function and malignant transformation, and epigenetic alterations are important causes of miRNAs dysregulation in cancer.Patched1 heterozygous (Ptch1+/-) mice, characterized by aberrant activation of the Sonic hedgehog (Shh) signaling pathway, are a well-known murine model of spontaneous and radiation-induced medulloblastoma (MB), a common pediatric brain tumor originating from neural granule cell progenitors (GCPs). The high sensitivity of neonatal Ptch1+/- mice to radiogenic MB is dependent on deregulation of the Ptch1 gene function. Ptch1 activates a growth and differentiation programme that is a strong candidate for regulation through the non-coding genome. Therefore we carried out miRNA next generation sequencing in ex vivo irradiated and control GCPs, isolated and purified from cerebella of neonatal WT and Ptch1+/- mice. We identified a subset of miRNAs, namely let-7 family and miR-17~92 cluster members, whose expression is altered in GCPs by radiation alone, or by synergistic interaction of radiation with Shh-deregulation. The same miRNAs were further validated in spontaneous and radiation-induced MBs from Ptch1+/- mice, confirming persistent deregulation of these miRNAs in the pathogenesis of MB.Our results support the hypothesis that miRNAs dysregulation is associated with radiosensitivity of GCPs and their neoplastic transformation in vivo.


Subject(s)
Cerebellar Neoplasms/genetics , Cerebellum/radiation effects , Medulloblastoma/genetics , MicroRNAs/genetics , Patched-1 Receptor/genetics , Transcriptome/radiation effects , Animals , Animals, Newborn , Cerebellum/metabolism , Cerebellum/pathology , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/radiation effects , Gene Regulatory Networks/radiation effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mice, Knockout , Patched-1 Receptor/metabolism , Signal Transduction/genetics , Signal Transduction/radiation effects
13.
Colloids Surf B Biointerfaces ; 148: 431-439, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27648774

ABSTRACT

Self-assembling plant virus nanoparticles (pVNPs) have started to be explored as nanometre-sized objects for biomedical applications, such as vaccine or drug delivery and imaging. Plant VNPs may be ideal tools in terms of biocompatibility and biodegradability endowed with a wide diversity of symmetries and dimensions, easy chemical/biological engineering, and rapid production in plants. Recently, we defined that icosahedral Tomato bushy stunt virus (TBSV) and filamentous Potato virus X (PVX) are neither toxic nor teratogenic. We report here the results of an interdisciplinary study aimed to define for the first time the biodistribution of unlabelled, unpegylated, underivatized TBSV and PVX by proved detecting antibodies. These data add new insights on the in vivo behaviour of these nano-objects and demonstrate that the pVNPs under scrutiny are each intrinsically endowed with peculiar properties foreshadowing different applications in molecular medicine.


Subject(s)
Genetic Vectors/pharmacokinetics , Nanoparticles/metabolism , Potexvirus/genetics , Tombusvirus/genetics , Virus Diseases/metabolism , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Brain/metabolism , Brain/virology , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Genome, Viral/genetics , Immunohistochemistry , Kidney/metabolism , Kidney/virology , Liver/metabolism , Liver/virology , Lung/metabolism , Lung/virology , Mice , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Potexvirus/immunology , Potexvirus/physiology , Spleen/metabolism , Spleen/virology , Time Factors , Tissue Distribution , Nicotiana/virology , Tombusvirus/immunology , Tombusvirus/physiology , Virion/genetics , Virion/physiology , Virus Diseases/virology
14.
Radiat Res ; 186(3): 315-21, 2016 09.
Article in English | MEDLINE | ID: mdl-27541824

ABSTRACT

While most of the evidence for radiation-induced late health effects relates to cancer, there has been increasing interest recently in the development of non-cancer diseases, including lens opacity, observed in populations exposed to low-dose radiation. In a recent study, we reported that mice heterozygous for the Patched1 (Ptch1) gene represented a novel and powerful animal model for this disorder, and a useful tool for investigating the mechanisms of radiogenic cataract development. Given the ongoing and considerable uncertainty in allowable lens dose levels and the existence of a threshold for the development of cataracts, we tested the effects of a decreasing range of radiation doses (2 Gy, 1 Gy and 0.5 Gy X rays) by irradiating groups of Ptch1(+/-) mice at 2 days of age. Our findings showed that at this dose range, acute exposure of this highly susceptible mouse model did not induce macroscopically detectable cataracts, and only the 2 Gy irradiated mice showed microscopic alterations of the lens. Molecular analyses performed to evaluate the induction of epithelial-mesenchymal transition (EMT) and subsequent fibrotic alterations in mouse lens cells also indicated the existence of a dose threshold for such effects in the mouse model used. The mechanisms of cataractogenesis remain unclear, and further experimental studies are essential to elucidate those mechanisms specific for cataract initiation and development after irradiation, as well as the underlying genetic factors controlling cataract susceptibility.


Subject(s)
Cataract/pathology , Nonlinear Dynamics , Patched-1 Receptor/deficiency , Radiation Injuries/pathology , Radiation Tolerance , Alleles , Animals , Cataract/etiology , Cataract/metabolism , Disease Models, Animal , Dose-Response Relationship, Radiation , Lens, Crystalline/pathology , Lens, Crystalline/radiation effects , Mice , Patched-1 Receptor/genetics , Radiation Injuries/etiology , Radiation Injuries/metabolism
15.
Oncotarget ; 6(34): 36098-112, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26452034

ABSTRACT

The hypothesis of transgenerational induction of increased cancer susceptibility after paternal radiation exposure has long been controversial because of inconsistent results and the lack of a mechanistic interpretation. Here, exploiting Ptch1 heterozygous knockout mice, susceptible to spontaneous and radiation-induced medulloblastoma, we show that exposure of paternal germ cells to 1 Gy X-rays, at the spermatogonial stage, increased by a considerable 1.4-fold the offspring susceptibility to medulloblastoma induced by neonatal irradiation. This effect gained further biological significance thanks to a number of supporting data on the immunohistochemical characterization of the target tissue and preneoplastic lesions (PNLs). These results altogether pointed to increased proliferation of cerebellar granule cell precursors and PNLs cells, which favoured the development of frank tumours. The LOH analysis of tumor DNA showed Ptch1 biallelic loss in all tumor samples, suggesting that mechanisms other than interstitial deletions, typical of radiation-induced medulloblastoma, did not account for the observed increased cancer risk. This data was supported by comet analysis showing no differences in DNA damage induction and repair in cerebellar cells as a function of paternal irradiation. Finally, we provide biological plausibility to our results offering evidence of a possible epigenetic mechanism of inheritance based on radiation-induced changes of the microRNA profile of paternal sperm.


Subject(s)
Cerebellar Neoplasms/etiology , Cerebellar Neoplasms/genetics , Medulloblastoma/etiology , Medulloblastoma/genetics , Neoplasms, Radiation-Induced/genetics , Receptors, Cell Surface/genetics , Animals , Cerebellar Neoplasms/pathology , Comet Assay , Female , Male , Medulloblastoma/pathology , Mice , Mice, Transgenic , Microarray Analysis , Patched Receptors , Patched-1 Receptor
16.
Oncotarget ; 6(31): 31263-71, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26359350

ABSTRACT

There is epidemiological evidence for increased non-cancer mortality, primarily due to circulatory diseases after radiation exposure above 0.5 Sv. We evaluated the effects of chronic low-dose rate versus acute exposures in a murine model of spontaneous atherogenesis. Female ApoE-/- mice (60 days) were chronically irradiated for 300 days with gamma rays at two different dose rates (1 mGy/day; 20 mGy/day), with total accumulated doses of 0.3 or 6 Gy. For comparison, age-matched ApoE-/- females were acutely exposed to the same doses and sacrificed 300 days post-irradiation. Mice acutely exposed to 0.3 or 6 Gy showed increased atherogenesis compared to age-matched controls, and this effect was persistent. When the same doses were delivered at low dose rate over 300 days, we again observed a significant impact on global development of atherosclerosis, although at 0.3 Gy effects were limited to the descending thoracic aorta. Our data suggest that a moderate dose of 0.3 Gy can have persistent detrimental effects on the cardiovascular system, and that a high dose of 6 Gy poses high risks at both high and low dose rates. Our results were clearly nonlinear with dose, suggesting that lower doses may be more damaging than predicted by a linear dose response.


Subject(s)
Aorta, Thoracic/radiation effects , Aortic Diseases/etiology , Apolipoproteins E/deficiency , Atherosclerosis/etiology , Radiation Dosage , Radiation Injuries, Experimental/etiology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Progression , Dose-Response Relationship, Radiation , Female , Linear Models , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic , Radiation Injuries, Experimental/genetics , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Risk Assessment , Time Factors
18.
Am J Pathol ; 185(1): 85-95, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25452120

ABSTRACT

Age-related cataract is the most common cause of visual impairment. Moreover, traumatic cataracts form after injury to the eye, including radiation damage. We report herein that sonic hedgehog (Shh) signaling plays a key role in cataract development and in normal lens response to radiation injury. Mice heterozygous for Patched 1 (Ptch1), the Shh receptor and negative regulator of the pathway, develop spontaneous cataract and are highly susceptible to cataract induction by exposure to ionizing radiation in early postnatal age, when lens epithelial cells undergo rapid expansion in the lens epithelium. Neonatally irradiated and control Ptch1(+/-) mice were compared for markers of progenitors, Shh pathway activation, and epithelial-to-mesenchymal transition (EMT). Molecular analyses showed increased expression of the EMT-related transforming growth factor ß/Smad signaling pathway in the neonatally irradiated lens, and up-regulation of mesenchymal markers Zeb1 and Vim. We further show a link between proliferation and the stemness property of lens epithelial cells, controlled by Shh. Our results suggest that Shh and transforming growth factor ß signaling cooperate to promote Ptch1-associated cataract development by activating EMT, and that the Nanog marker of pluripotent cells may act as the primary transcription factor on which both signaling pathways converge after damage. These findings highlight a novel function of Shh signaling unrelated to cancer and provide a new animal model to investigate the molecular pathogenesis of cataract formation.


Subject(s)
Cataract/metabolism , Gene Expression Regulation , Lens, Crystalline/metabolism , Receptors, Cell Surface/genetics , Alleles , Animals , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Profiling , Hedgehog Proteins/metabolism , Heterozygote , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lens, Crystalline/pathology , Lens, Crystalline/radiation effects , Mice , Mice, Transgenic , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Vimentin/metabolism , X-Rays , Zinc Finger E-box-Binding Homeobox 1
19.
Int J Nanomedicine ; 9: 1919-29, 2014.
Article in English | MEDLINE | ID: mdl-24790434

ABSTRACT

Superparamagnetic iron oxide nanoparticles are candidate contrast agents for magnetic resonance imaging and targeted drug delivery. Biodistribution and toxicity assessment are critical for the development of nanoparticle-based drugs, because of nanoparticle-enhanced biological reactivity. Here, we investigated the uptake, in vivo biodistribution, and in vitro and in vivo potential toxicity of manganese ferrite (MnFe2O4) nanoparticles, synthesized by an original high-yield, low-cost mechanochemical process. Cultures of murine Balb/3T3 fibroblasts were exposed for 24, 48, or 72 hours to increasing ferrofluid concentrations. Nanoparticle cellular uptake was assessed by flow-cytometry scatter-light measurements and microscopy imaging after Prussian blue staining; cytotoxicity was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony-forming assays. After a single intravenous injection, in vivo nanoparticle biodistribution and clearance were evaluated in mice by Mn spectrophotometric determination and Prussian blue staining in the liver, kidneys, spleen, and brain at different posttreatment times up to 21 days. The same organs were analyzed for any possible histopathological change. The in vitro study demonstrated dose-dependent nanoparticle uptake and statistically significant cytotoxic effects from a concentration of 50 µg/mL for the MTT assay and 20 µg/mL for the colony-forming assay. Significant increases in Mn concentrations were detected in all analyzed organs, peaking at 6 hours after injection and then gradually declining. Clearance appeared complete at 7 days in the kidneys, spleen, and brain, whereas in the liver Mn levels remained statistically higher than in vehicle-treated mice up to 3 weeks postinjection. No evidence of irreversible histopathological damage to any of the tested organs was observed. A comparison of the lowest in vitro toxic concentration with the intravenously injected dose and the administered dose of other ferrofluid drugs currently in clinical practice suggests that there might be sufficient safety margins for further development of our formulation.


Subject(s)
Cell Survival/drug effects , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/toxicity , Manganese/chemistry , Manganese/toxicity , Animals , BALB 3T3 Cells , Colloids/chemical synthesis , Colloids/toxicity , Contrast Media , Diffusion , Dose-Response Relationship, Drug , Drug Compounding/methods , Female , Lethal Dose 50 , Materials Testing , Mice , Organ Specificity , Solutions , Stress, Mechanical , Survival Rate , Tissue Distribution
20.
Redox Biol ; 2: 580-9, 2014.
Article in English | MEDLINE | ID: mdl-24688894

ABSTRACT

N(6)-isopentenyladenosine (i(6)A), a naturally occurring modified nucleoside, inhibits the proliferation of human tumor cell lines in vitro, but its mechanism of action remains unclear. Treatment of MCF7 human breast adenocarcinoma cells with i(6)A or with three synthetic analogs (allyl(6)A, benzyl(6)A, and butyl(6)A) inhibited growth and altered gene expression. About 60% of the genes that were differentially expressed in response to i(6)A treatment were also modulated by the analogs, and pathway enrichment analysis identified the NRF2-mediated oxidative stress response as being significantly modulated by all four compounds. Luciferase reporter gene assays in transfected MCF7 cells confirmed that i(6)A activates the transcription factor NRF2. Assays for cellular production of reactive oxygen species indicated that i(6)A and analogs had antioxidant effects, reducing basal levels and inhibiting the H2O2- or 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced production in MCF7 or dHL-60 (HL-60 cells induced to differentiate along the neutrophilic lineage) cell lines, respectively. In vivo, topical application of i(6)A or benzyl(6)A to mouse ears prior to TPA stimulation lessened the inflammatory response and significantly reduced the number of infiltrating neutrophils. These results suggest that i(6)A and analogs trigger a cellular response against oxidative stress and open the possibility of i(6)A and benzyl(6)A being used as topical anti-inflammatory drugs.


Subject(s)
Antioxidants/administration & dosage , Isopentenyladenosine/analogs & derivatives , Isopentenyladenosine/administration & dosage , NF-E2-Related Factor 2/metabolism , Otitis/drug therapy , Administration, Topical , Animals , Antioxidants/pharmacology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , HL-60 Cells , Humans , Hydrogen Peroxide/adverse effects , Isopentenyladenosine/pharmacology , MCF-7 Cells , Mice , Otitis/chemically induced , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tetradecanoylphorbol Acetate/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...