Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cells ; 13(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891031

ABSTRACT

Apolipoprotein E (ApoE) is a lipid carrier in both the peripheral and the central nervous systems (CNSs). Lipid-loaded ApoE lipoprotein particles bind to several cell surface receptors to support membrane homeostasis and brain injury repair. In the brain, ApoE is produced predominantly by astrocytes, but it is also abundantly expressed in most neurons of the CNS. In this study, we addressed the role of ApoE in the hippocampus in mice, focusing on its role in response to radiation injury. To this aim, 8-week-old, wild-type, and ApoE-deficient (ApoE-/-) female mice were acutely whole-body irradiated with 3 Gy of X-rays (0.89 Gy/min), then sacrificed 150 days post-irradiation. In addition, age-matching ApoE-/- females were chronically whole-body irradiated (20 mGy/d, cumulative dose of 3 Gy) for 150 days at the low dose-rate facility at the Institute of Environmental Sciences (IES), Rokkasho, Japan. To seek for ApoE-dependent modification during lineage progression from neural stem cells to neurons, we have evaluated the cellular composition of the dentate gyrus in unexposed and irradiated mice using stage-specific markers of adult neurogenesis. Our findings indicate that ApoE genetic inactivation markedly perturbs adult hippocampal neurogenesis in unexposed and irradiated mice. The effect of ApoE inactivation on the expression of a panel of miRNAs with an established role in hippocampal neurogenesis, as well as its transcriptional consequences in their target genes regulating neurogenic program, have also been analyzed. Our data show that the absence of ApoE-/- also influences synaptic functionality and integration by interfering with the regulation of mir-34a, mir-29b, and mir-128b, leading to the downregulation of synaptic markers PSD95 and synaptophysin mRNA. Finally, compared to acute irradiation, chronic exposure of ApoE null mice yields fewer consequences except for the increased microglia-mediated neuroinflammation. Exploring the function of ApoE in the hippocampus could have implications for developing therapeutic approaches to alleviate radiation-induced brain injury.


Subject(s)
Apolipoproteins E , Hippocampus , MicroRNAs , Radiation, Ionizing , Animals , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hippocampus/metabolism , Hippocampus/radiation effects , Mice , Female , MicroRNAs/metabolism , MicroRNAs/genetics , Mice, Inbred C57BL , Neurons/metabolism , Neurons/radiation effects , Neurogenesis/radiation effects , Whole-Body Irradiation , Radiation Exposure/adverse effects , Dentate Gyrus/metabolism , Dentate Gyrus/radiation effects , Dentate Gyrus/pathology
2.
Cancers (Basel) ; 15(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37444528

ABSTRACT

Recent epidemiologic studies support an association between chronic low-dose radiation exposure and the development of cardiovascular disease (CVD). The molecular mechanisms underlying the adverse effect of chronic low dose exposure are not fully understood. To address this issue, we have investigated changes in the heart proteome of ApoE deficient (ApoE-/-) C57Bl/6 female mice chronically irradiated for 300 days at a very low dose rate (1 mGy/day) or at a low dose rate (20 mGy/day), resulting in cumulative whole-body doses of 0.3 Gy or 6.0 Gy, respectively. The heart proteomes were compared to those of age-matched sham-irradiated ApoE-/- mice using label-free quantitative proteomics. Radiation-induced proteome changes were further validated using immunoblotting, enzyme activity assays, immunohistochemistry or targeted transcriptomics. The analyses showed persistent alterations in the cardiac proteome at both dose rates; however, the effect was more pronounced following higher dose rates. The altered proteins were involved in cardiac energy metabolism, ECM remodelling, oxidative stress, and ageing signalling pathways. The changes in PPARα, SIRT, AMPK, and mTOR signalling pathways were found at both dose rates and in a dose-dependent manner, whereas more changes in glycolysis and ECM remodelling were detected at the lower dose rate. These data provide strong evidence for the possible risk of cardiac injury following chronic low dose irradiation and show that several affected pathways following chronic irradiation overlap with those of ageing-associated heart pathology.

3.
Int J Mol Sci ; 24(10)2023 May 15.
Article in English | MEDLINE | ID: mdl-37240150

ABSTRACT

Pelvic radiation disease (PRD), a frequent side effect in patients with abdominal/pelvic cancers treated with radiotherapy, remains an unmet medical need. Currently available preclinical models have limited applications for the investigation of PRD pathogenesis and possible therapeutic strategies. In order to select the most effective irradiation protocol for PRD induction in mice, we evaluated the efficacy of three different locally and fractionated X-ray exposures. Using the selected protocol (10 Gy/day × 4 days), we assessed PRD through tissue (number and length of colon crypts) and molecular (expression of genes involved in oxidative stress, cell damage, inflammation, and stem cell markers) analyses at short (3 h or 3 days after X-ray) and long (38 days after X-rays) post-irradiation times. The results show that a primary damage response in term of apoptosis, inflammation, and surrogate markers of oxidative stress was found, thus determining a consequent impairment of cell crypts differentiation and proliferation as well as a local inflammation and a bacterial translocation to mesenteric lymph nodes after several weeks post-irradiation. Changes were also found in microbiota composition, particularly in the relative abundance of dominant phyla, related families, and in alpha diversity indices, as an indication of dysbiotic conditions induced by irradiation. Fecal markers of intestinal inflammation, measured during the experimental timeline, identified lactoferrin, along with elastase, as useful non-invasive tools to monitor disease progression. Thus, our preclinical model may be useful to develop new therapeutic strategies for PRD treatment.


Subject(s)
Radiation Injuries , Mice , Animals , X-Rays , Disease Models, Animal , Apoptosis/radiation effects , Inflammation
4.
Mol Neurobiol ; 58(4): 1634-1649, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33230715

ABSTRACT

We here characterize the response to the extremely low-frequency (ELF) magnetic field (MF, 50 Hz, 1 mT) of SH-SY5Y human neuroblastoma cells, cultured in a three-dimensional (3D) Alvetex® scaffold compared to conventional two-dimensional (2D) monolayers. We proved that the growing phenotype of proliferating SH-SY5Y cells is not affected by the culturing conditions, as morphology, cell cycle distribution, proliferation/differentiation gene expression of 3D-cultures overlap what reported in 2D plates. In response to 72-h exposure to 50-Hz MF, we demonstrated that no proliferation change and apoptosis activation occur in both 2D and 3D cultures. Consistently, no modulation of Ki67, MYCN, CCDN1, and Nestin, of invasiveness and neo-angiogenesis-controlling genes (HIF-1α, VEGF, and PDGF) and of microRNA epigenetic signature (miR-21-5p, miR-222-3p and miR-133b) is driven by ELF exposure. Conversely, intracellular glutathione content and SOD1 expression are exclusively impaired in 3D-culture cells in response to the MF, whereas no change of such redox modulators is observed in SH-SY5Y cells if grown on 2D monolayers. Moreover, ELF-MF synergizes with the differentiating agents to stimulate neuroblastoma differentiation into a dopaminergic (DA) phenotype in the 3D-scaffold culture only, as growth arrest and induction of p21, TH, DAT, and GAP43 are reported in ELF-exposed SH-SY5Y cells exclusively if grown on 3D scaffolds. As overall, our findings prove that 3D culture is a more reliable experimental model for studying SH-SY5Y response to ELF-MF if compared to 2D conventional monolayer, and put the bases for promoting 3D systems in future studies addressing the interaction between electromagnetic fields and biological systems.


Subject(s)
Cell Culture Techniques , Magnetic Fields , Neuroblastoma/pathology , Apoptosis , Biomarkers/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Dopaminergic Neurons/pathology , Glutathione/deficiency , Glutathione/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neovascularization, Physiologic , Neuroblastoma/genetics , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
6.
J Proteome Res ; 17(4): 1677-1689, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29560722

ABSTRACT

High-dose ionizing radiation is known to induce adverse effects such as inflammation and fibrosis in the heart. Transcriptional regulators PPARα and TGFß are known to be involved in this radiation response. PPARα, an anti-inflammatory transcription factor controlling cardiac energy metabolism, is inactivated by irradiation. The pro-inflammatory and pro-fibrotic TGFß is activated by irradiation via SMAD-dependent and SMAD-independent pathways. The goal of this study was to investigate how altering the level of PPARα influences the radiation response of these signaling pathways. For this purpose, we used genetically modified C57Bl/6 mice with wild type (+/+), heterozygous (+/-) or homozygous (-/-) PPARα genotype. Mice were locally irradiated to the heart using doses of 8 or 16 Gy; the controls were sham-irradiated. The heart tissue was investigated using label-free proteomics 20 weeks after the irradiation and the predicted pathways were validated using immunoblotting, ELISA, and immunohistochemistry. The heterozygous PPARα mice showed most radiation-induced changes in the cardiac proteome, whereas the homozygous PPARα mice showed the least changes. Irradiation induced SMAD-dependent TGFß signaling independently of the PPARα status, but the presence of PPARα was necessary for the activation of the SMAD-independent pathway. These data indicate a central role of PPARα in cardiac response to ionizing radiation.


Subject(s)
Heart/radiation effects , Myocardium/metabolism , PPAR alpha/physiology , Transforming Growth Factor beta/metabolism , Animals , Genotype , Heterozygote , Mice , Mice, Inbred C57BL , Myocardium/chemistry , PPAR alpha/genetics , Proteomics , Signal Transduction , Smad Proteins/metabolism
7.
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.

8.
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
9.
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
10.
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
11.
Mol Cancer Ther ; 15(6): 1177-89, 2016 06.
Article in English | MEDLINE | ID: mdl-26960983

ABSTRACT

Aberrant activation of the Hedgehog (Hh) signaling pathway is implicated in the pathogenesis of many cancers, including medulloblastoma and basal cell carcinoma (BCC). In this study, using neonatally irradiated Ptch1(+/-) mice as a model of Hh-dependent tumors, we investigated the in vivo effects of MK-4101, a novel SMO antagonist, for the treatment of medulloblastoma and BCC. Results clearly demonstrated a robust antitumor activity of MK-4101, achieved through the inhibition of proliferation and induction of extensive apoptosis in tumor cells. Of note, beside antitumor activity on transplanted tumors, MK-4101 was highly efficacious against primary medulloblastoma and BCC developing in the cerebellum and skin of Ptch1(+/-) mice. By identifying the changes induced by MK-4101 in gene expression profiles in tumors, we also elucidated the mechanism of action of this novel, orally administrable compound. MK-4101 targets the Hh pathway in tumor cells, showing the maximum inhibitory effect on Gli1 MK-4101 also induced deregulation of cell cycle and block of DNA replication in tumors. Members of the IGF and Wnt signaling pathways were among the most highly deregulated genes by MK-4101, suggesting that the interplay among Hh, IGF, and Wnt is crucial in Hh-dependent tumorigenesis. Altogether, the results of this preclinical study support a therapeutic opportunity for MK-4101 in the treatment of Hh-driven cancers, also providing useful information for combination therapy with drugs targeting pathways cooperating with Hh oncogenic activity. Mol Cancer Ther; 15(6); 1177-89. ©2016 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Carcinoma, Basal Cell/drug therapy , Cerebellar Neoplasms/drug therapy , Hedgehog Proteins/antagonists & inhibitors , Isoxazoles/administration & dosage , Isoxazoles/chemical synthesis , Medulloblastoma/drug therapy , Triazoles/administration & dosage , Triazoles/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cerebellar Neoplasms/metabolism , Humans , Isoxazoles/pharmacology , Medulloblastoma/metabolism , Mice , Neoplasm Transplantation , Random Allocation , Signal Transduction/drug effects , Triazoles/pharmacology , Xenograft Model Antitumor Assays
12.
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
13.
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
14.
Stem Cells ; 31(11): 2506-16, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23897709

ABSTRACT

Neural stem cells are highly susceptible to radiogenic DNA damage, however, little is known about their mechanisms of DNA damage response (DDR) and the long-term consequences of genotoxic exposure. Patched1 heterozygous mice (Ptc1(+/-)) provide a powerful model of medulloblastoma (MB), a frequent pediatric tumor of the cerebellum. Irradiation of newborn Ptc1(+/-) mice dramatically increases the frequency and shortens the latency of MB. In this model, we investigated the mechanisms through which multipotent neural progenitors (NSCs) and fate-restricted progenitor cells (PCs) of the cerebellum respond to DNA damage induced by radiation, and the long-term developmental and oncogenic consequences. These responses were assessed in mice exposed to low (0.25 Gy) or high (3 Gy) radiation doses at embryonic day 13.5 (E13.5), when NSCs giving rise to the cerebellum are specified but the external granule layer (EGL) has not yet formed, or at E16.5, during the expansion of granule PCs to form the EGL. We found crucial differences in DDR and apoptosis between NSCs and fate-restricted PCs, including lack of p21 expression in NSCs. NSCs also appear to be resistant to oncogenesis from low-dose radiation exposure but more vulnerable at higher doses. In addition, the pathway to DNA repair and the pattern of oncogenic alterations were strongly dependent on age at exposure, highlighting a differentiation-stage specificity of DNA repair pathways in NSCs and PCs. These findings shed light on the mechanisms used by NSCs and PCs to maintain genome integrity during neurogenesis and may have important implications for radiation risk assessment and for development of targeted therapies against brain tumors.


Subject(s)
Cerebellum/growth & development , Cerebellum/radiation effects , Neural Stem Cells/radiation effects , Stem Cells/physiology , Stem Cells/radiation effects , Animals , Apoptosis/radiation effects , Cell Differentiation/physiology , Cell Differentiation/radiation effects , Cerebellum/cytology , Cerebellum/pathology , DNA Damage , DNA Repair , Medulloblastoma/genetics , Medulloblastoma/pathology , Medulloblastoma/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Stem Cells/cytology
15.
Int J Radiat Oncol Biol Phys ; 86(5): 993-9, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23755921

ABSTRACT

PURPOSE: To investigate the tissue dependence in transmission of abscopal radiation signals and their oncogenic consequences in a radiosensitive mouse model and to explore the involvement of gap junction intercellular communication (GJIC) in mediating radiation tumorigenesis in off-target mouse skin. METHODS AND MATERIALS: Patched1 heterozygous (Ptch1(+/-)) mice were irradiated at postnatal day 2 (P2) with 10 Gy of x-rays. Individual lead cylinders were used to protect the anterior two-thirds of the body, whereas the hindmost part was directly exposed to radiation. To test the role of GJICs and their major constituent connexin43 (Cx43), crosses between Ptch1(+/-) and Cx43(+/-) mice were similarly irradiated. These mouse groups were monitored for their lifetime, and skin basal cell carcinomas (BCCs) were counted and recorded. Early responses to DNA damage - Double Strand Breaks (DSBs) and apoptosis - were also evaluated in shielded and directly irradiated skin areas. RESULTS: We report abscopal tumor induction in the shielded skin of Ptch1(+/-) mice after partial-body irradiation. Endpoints were induction of early nodular BCC-like tumors and macroscopic infiltrative BCCs. Abscopal tumorigenesis was significantly modulated by Cx43 status, namely, Cx43 reduction was associated with decreased levels of DNA damage and oncogenesis in out-of-field skin, suggesting a key role of GJIC in transmission of oncogenic radiation signals to unhit skin. CONCLUSIONS: Our results further characterize the nature of abscopal responses and the implications they have on pathologic processes in different tissues, including their possible underlying mechanistic bases.


Subject(s)
Carcinoma, Basal Cell/etiology , Gap Junctions/physiology , Neoplasms, Radiation-Induced/etiology , Radiation Tolerance/physiology , Skin Neoplasms/etiology , Skin/radiation effects , Animals , Apoptosis , Carcinoma, Basal Cell/pathology , Carcinoma, Basal Cell/physiopathology , Connexin 43/genetics , Connexin 43/physiology , Crosses, Genetic , DNA Damage , Gene Knockdown Techniques , Mice , Neoplasms, Radiation-Induced/pathology , Neoplasms, Radiation-Induced/physiopathology , Patched Receptors , Patched-1 Receptor , Radiation Protection/methods , Receptors, Cell Surface/genetics , Skin/pathology , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology
16.
Int J Radiat Oncol Biol Phys ; 85(3): 813-9, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-22975608

ABSTRACT

PURPOSE: To investigate the dose and spatial dependence of abscopal radiation effects occurring in vivo in the mouse, along with their tumorigenic potential in the central nervous system (CNS) of a radiosensitive mouse model. METHODS AND MATERIALS: Patched1 (Ptch1)(+/-) mice, carrying a germ-line heterozygous inactivating mutation in the Ptch1 gene and uniquely susceptible to radiation damage in neonatal cerebellum, were exposed directly to ionizing radiation (1, 2, or 3 Gy of x-rays) or treated in a variety of partial-body irradiation protocols, in which the animals' head was fully protected by suitable lead cylinders while the rest of the body was exposed to x-rays in full or in part. Apoptotic cell death was measured in directly irradiated and shielded cerebellum shortly after irradiation, and tumor development was monitored in lifetime groups. The same endpoints were measured using different shielding geometries in mice irradiated with 3 or 10 Gy of x-rays. RESULTS: Although dose-dependent cell death was observed in off-target cerebellum for all doses and shielding conditions tested, a conspicuous lack of abscopal response for CNS tumorigenesis was evident at the lowest dose of 1 Gy. By changing the amount of exposed body volume, the shielding geometry could also significantly modulate tumorigenesis depending on dose. CONCLUSIONS: We conclude that interplay between radiation dose and exposed tissue volume plays a critical role in nontargeted effects occurring in mouse CNS under conditions relevant to humans. These findings may help understanding the mechanisms of long-range radiation signaling in harmful effects, including carcinogenesis, occurring in off-target tissues.


Subject(s)
Bystander Effect/physiology , Cerebellar Neoplasms/etiology , Cerebellum/radiation effects , Neoplasms, Radiation-Induced/etiology , Radiation Protection/methods , Radiation Tolerance , Animals , Cell Death/physiology , Dose-Response Relationship, Radiation , Germ-Line Mutation/genetics , Mice , Patched Receptors , Patched-1 Receptor , Radiation Tolerance/genetics , Receptors, Cell Surface/genetics , Time Factors , Whole-Body Irradiation/methods
17.
Cancer Lett ; 308(2): 197-202, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21636213

ABSTRACT

Gender-related differences in medulloblastoma (MB) development have been reported with a higher incidence in males (slightly above 60%) than in females, female gender being also a significantly favorable prognostic factor in MB. The present study focused on the evaluation of the mechanisms by which estrogens protect against MB formation. To this end, we used a well characterized mouse model of MB - the Patched1 heterozygous mice. Ovariectomized mice were treated with 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN), a highly potent ERß agonist, or 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT), a highly potent ERα agonist. Our results show that the ERß selective agonist DPN significantly inhibits development of MB preneoplastic lesions when compared with untreated ovariectomized mice, restoring the final incidence to that observed in the intact controls, and that these effects were achieved via activation of anti-proliferative and pro-apototic pathways. On the other hand, the ERα selective agonist PPT did not influence MB tumorigenesis relative to untreated ovariectomized mice.


Subject(s)
Estrogen Receptor beta/agonists , Medulloblastoma/pathology , Nitriles/pharmacology , Propionates/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Nitriles/therapeutic use , Propionates/therapeutic use , Signal Transduction/drug effects
18.
Mutagenesis ; 26(4): 563-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21498842

ABSTRACT

Male germ cells have been shown to differ in their DNA damage response (DDR) with respect to somatic cells. In addition, DDR pathways are modulated along spermatogenesis, accompanying profound chromatin modifications. Histone H2AX phosphorylation is a fundamental step of DDR. Few data are available on the long-term kinetics of phosphorylated H2AX (γ-H2AX) after in vivo irradiation. We have investigated, by microscopic and flow cytometric immunochemistry, γ-H2AX induction and removal in testicular cells of irradiated mice, in comparison with bone marrow cells. In unirradiated testicular cells, much higher levels of γ-H2AX were measured by flow cytometry with respect to bone marrow cells. Irradiation induced a redistribution of γ-H2AX into discrete foci detectable by microscopy. In irradiated bone marrow, the percentage of labelled cells peaked at 1 h and rapidly declined, in agreement with data on in vitro cell lines. In contrast, spermatocytes and round spermatids showed persistent labelling until 48 h. During this time, in spermatids, topological changes were observed in γ-H2AX foci from a pattern of many uncountable dots to a pattern of few large spots. Observations of testicular sections confirmed this trend in the reduction of foci number in spite of substantially invariable percentages of labelled cells in the analysed timeframe. To assess whether γ-H2AX persistence in testicular cells was due to unrepaired DNA breaks, we performed comet assay and immunofluorescence analysis of Mdc1, a marker of DDR different from γ-H2AX. Comet assay showed that most breaks were repaired within 2 h. Forty-eight hours after irradiation, contrary to γ-H2AX foci that remained detectable in 80% of initially labelled cells, Mdc1 foci were observed in only 20-30% of cells. These data suggest that, at long times after irradiation, mechanisms additional to impairment of DNA break repair may account for the long persistence of γ-H2AX foci in male germ cells.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Histones/metabolism , Testis/metabolism , Testis/radiation effects , Animals , Comet Assay , Flow Cytometry , Immunohistochemistry , Kinetics , Male , Mice , Mice, Inbred C57BL , Phosphorylation/radiation effects , Testis/pathology , X-Rays
19.
Mol Cancer ; 9: 53, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20214787

ABSTRACT

BACKGROUND: Medulloblastoma is amongst the most common malignant brain tumors in childhood, arising from neoplastic transformation of granule neuron precursors (GNPs) of the cerebellum via deregulation of pathways involved in cerebellar development. Deregulation of the Sonic hedgehog/Patched1 (Shh/Ptc1) signaling pathway predisposes humans and mice to medulloblastoma. In the brain, insulin-like growth factor (IGF-I) plays a critical role during development as a neurotrophic and neuroprotective factor, and in tumorigenesis, as IGF-I receptor is often activated in medulloblastomas. RESULTS: To investigate the mechanisms of genetic interactions between Shh and IGF signaling in the cerebellum, we crossed nestin/IGF-I transgenic (IGF-I Tg) mice, in which transgene expression occurs in neuron precursors, with Ptc1+/- knockout mice, a model of medulloblastoma in which cancer develops in a multistage process. The IGF-I transgene produced a marked brain overgrowth, and significantly accelerated tumor development, increasing the frequency of pre-neoplastic lesions as well as full medulloblastomas in Ptc1+/-/IGF-I Tg mice. Mechanistically, tumor promotion by IGF-I mainly affected preneoplastic stages through de novo formation of lesions, while not influencing progression rate to full tumors. We also identified a marked increase in survival and proliferation, and a strong suppression of differentiation in neural precursors. CONCLUSIONS: As a whole, our findings indicate that IGF-I overexpression in neural precursors leads to brain overgrowth and fosters external granular layer (EGL) proliferative lesions through a mechanism favoring proliferation over terminal differentiation, acting as a landscape for tumor growth. Understanding the molecular events responsible for cerebellum development and their alterations in tumorigenesis is critical for the identification of potential therapeutic targets.


Subject(s)
Cerebellum/embryology , Cerebellum/pathology , Insulin-Like Growth Factor I/genetics , Precancerous Conditions/embryology , Precancerous Conditions/pathology , Receptors, Cell Surface/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cerebellum/metabolism , Humans , Intermediate Filament Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nestin , Neurons/metabolism , Neurons/pathology , Organ Size , Patched Receptors , Patched-1 Receptor , Phenotype , Promoter Regions, Genetic/genetics , Receptors, Cell Surface/genetics , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Transgenes/genetics
20.
Int J Cancer ; 127(12): 2749-57, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21351254

ABSTRACT

Medulloblastoma (MB) is the most common pediatric tumor of the CNS, representing ∼20% of all childhood CNS tumors. Although in recent years many molecular mechanisms that control MB development have been clarified, the effects of biological factors such as sex on this tumor remain to be explained. Epidemiological data, in fact, indicate a significant difference in the incidence of MB between the 2 sexes, with considerably higher susceptibility of males than females. Besides this different susceptibility, female sex is also a significant favorable prognostic factor in MB, with girls having a much better outcome. Despite these literature data, there has been little investigation into estrogen influence on MB development. In our study, we evaluated how hormone deficiency resulting from ovariectomy and hormone replacement influences the development of early and advanced MB stages in Patched1 heterozygous mice, a well-characterized mouse model of radiation-induced MB. Susceptibility to MB development was significantly increased in ovariectomized Ptch1(+/-) females and restored to levels observed in control mice after estrogen replacement. We next investigated the molecular mechanisms by which estrogen might influence tumor progression and show that ERß, but not ERα, is involved in modulation of MB development by estrogens. Finally, our study shows that a functional interaction between estrogen- and IGF-I-mediated pathways may be responsible for the effects observed.


Subject(s)
Cerebellar Neoplasms/prevention & control , Estradiol/therapeutic use , Estrogens/therapeutic use , Medulloblastoma/prevention & control , Receptors, Cell Surface/physiology , Animals , Blotting, Western , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Estrogen Receptor beta/metabolism , Female , Heterozygote , Immunoenzyme Techniques , Insulin-Like Growth Factor I/metabolism , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Ovariectomy , Patched Receptors , Patched-1 Receptor , RNA, Messenger/genetics , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...