Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
3.
Radiat Environ Biophys ; 61(4): 485-504, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36001144

RESUMEN

Partial body exposure and inhomogeneous dose delivery are features of the majority of medical and occupational exposure situations. However, mounting evidence indicates that the effects of partial body exposure are not limited to the irradiated area but also have systemic effects that are propagated outside the irradiated field. It was the aim of the "Partial body exposure" session within the MELODI workshop 2020 to discuss recent developments and insights into this field by covering clinical, epidemiological, dosimetric as well as mechanistic aspects. Especially the impact of out-of-field effects on dysfunctions of immune cells, cardiovascular diseases and effects on the brain were debated. The presentations at the workshop acknowledged the relevance of out-of-field effects as components of the cellular and organismal radiation response. Furthermore, their importance for the understanding of radiation-induced pathologies, for the discovery of early disease biomarkers and for the identification of high-risk organs after inhomogeneous exposure was emphasized. With the rapid advancement of clinical treatment modalities, including new dose rates and distributions a better understanding of individual health risk is urgently needed. To achieve this, a deeper mechanistic understanding of out-of-field effects in close connection to improved modelling was suggested as priorities for future research. This will support the amelioration of risk models and the personalization of risk assessments for cancer and non-cancer effects after partial body irradiation.


Asunto(s)
Enfermedades Cardiovasculares , Radiometría , Humanos , Medición de Riesgo
4.
Radiat Res ; 197(1): 36-42, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33652474

RESUMEN

Epidemiological studies suggest an increased incidence and risk of cataract after low-dose (<2 Gy) ionizing radiation exposures. However, the biological mechanism(s) of this process are not fully understood. DNA damage and repair are thought to have a contributing role in radiation-induced cataractogenesis. Recently we have reported an inverse dose-rate effect, as well as the low-dose response, of DNA damage and repair in lens epithelial cells (LECs). Here, we present further initial findings from two mutated strains (Ercc2+/- and Ptch1+/-) of mice, both reportedly susceptible to radiation-induced cataract, and their DNA damage and repair response to low-dose and low-dose-rate gamma rays. Our results support the hypothesis that the lens epithelium responds differently to radiation than other tissues, with reported radiation susceptibility to DNA damage not necessarily translating to the LECs. Genetic predisposition and strain(s) of mice have a significant role in radiation-induced cataract susceptibility.


Asunto(s)
Catarata/etiología , Daño del ADN/efectos de la radiación , Cristalino/efectos de los fármacos , Animales , Células Epiteliales , Rayos gamma , Humanos , Ratones , Receptor Patched-1/metabolismo , Exposición a la Radiación , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo
5.
Radiat Res ; 197(1): 43-56, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33857285

RESUMEN

Experimental mouse studies are important to gain a comprehensive, quantitative and mechanistic understanding of the biological factors that modify individual risk of radiation-induced health effects, including age at exposure, dose, dose rate, organ/tissue specificity and genetic factors. In this study, neonatal Ptch1+/- mice bred on CD1 and C57Bl/6 background received whole-body irradiation at postnatal day 2. This time point represents a critical phase in the development of the eye lens, cerebellum and dentate gyrus (DG), when they are also particularly susceptible to radiation effects. Irradiation was performed with γ rays (60Co) at doses of 0.5, 1 and 2 Gy, delivered at 0.3 Gy/min or 0.063 Gy/min. Wild-type and mutant mice were monitored for survival, lens opacity, medulloblastoma (MB) and neurogenesis defects. We identified an inverse genetic background-driven relationship between the radiosensitivity to induction of lens opacity and MB and that to neurogenesis deficit in Ptch1+/- mutants. In fact, high incidence of radiation-induced cataract and MB were observed in Ptch1+/-/CD1 mutants that instead showed no consequence of radiation exposure on neurogenesis. On the contrary, no induction of radiogenic cataract and MB was reported in Ptch1+/-/C57Bl/6 mice that were instead susceptible to induction of neurogenesis defects. Compared to Ptch1+/-/CD1, the cerebellum of Ptch1+/-/C57Bl/6 mice showed increased radiosensitivity to apoptosis, suggesting that differences in processing radiation-induced DNA damage may underlie the opposite strain-related radiosensitivity to cancer and non-cancer pathologies. Altogether, our results showed lack of dose-rate-related effects and marked influence of genetic background on the radiosensitivity of Ptch1+/-mice, supporting a major contribution of individual sensitivity to radiation risk in the population.


Asunto(s)
Meduloblastoma/etnología , Neoplasias Inducidas por Radiación/etiología , Animales , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Antecedentes Genéticos , Humanos , Cristalino/efectos de la radiación , Ratones Endogámicos C57BL , Neurogénesis , Tolerancia a Radiación , Irradiación Corporal Total
6.
Radiat Res ; 197(1): 22-35, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33857324

RESUMEN

One harmful long-term effect of ionizing radiation is cataract development. Recent studies have been focused on elucidating the mechanistic pathways involved in this pathogenesis. Since accumulating evidence has established a role of microRNAs in ocular diseases, including cataract, the goal of this work was to determine the microRNA signature of the mouse lens, at short time periods postirradiation, to understand the mechanisms related to radio-induced cataractogenesis. To evaluate the differences in the microRNA profiles, 10-week-old Patched1 heterozygous (Ptch1+/-) mice, bred onto two different genetic backgrounds (CD1 and C57Bl/6J), received whole-body 2 Gy γ-ray irradiation, and 24 h later lenses were collected. Next-generation sequencing and bioinformatics analysis revealed that genetic background markedly influenced the list of the deregulated microRNAs and the mainly predicted perturbed biological functions of 2 Gy irradiated Ptch1+/- mouse lenses. We identified a subset of microRNAs with a contra-regulated expression between strains, with a key role in regulating Toll-like receptor (TLR)-signaling pathways. Furthermore, a detailed analysis of miRNome data showed a completely different DNA damage response in mouse lenses 24 h postirradiation, mainly mediated by a marked upregulation of p53 signaling in Ptch1+/-/C57Bl/6J lenses that was not detected on a CD1 background. We propose a strict interplay between p53 and TLR signaling in Ptch1+/-/C57Bl/6J lenses shortly after irradiation that could explain both the resistance of this strain to developing lens opacities and the susceptibility of CD1 background to radiation-induced cataractogenesis through activation of epithelial-mesenchymal transition.


Asunto(s)
Catarata/etiología , Cristalino/efectos de la radiación , Animales , Daño del ADN/efectos de la radiación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Antecedentes Genéticos , Humanos , Ratones Endogámicos C57BL , MicroARNs , Receptor Patched-1/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Irradiación Corporal Total
7.
Radiat Res ; 197(1): 57-66, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33984859

RESUMEN

Recent epidemiological findings and reanalysis of historical data suggest lens opacities resulting from ionizing radiation exposures are likely induced at lower doses than previously thought. These observations have led to ICRP recommendations for a reduction in the occupational dose limits for the eye lens, as well as subsequent implementation in EU member states. The EU CONCERT LDLensRad project was initiated to further understand the effects of ionizing radiation on the lens and identify the mechanism(s) involved in radiation-induced cataract, as well as the impact of dose and dose-rate. Here, we present the results of a long-term study of changes to lens opacity in male and female adult mice from a variety of different genetic (radiosensitive or radioresistant) backgrounds, including mutant strains Ercc2 and Ptch1, which were assumed to be susceptible to radiation-induced lens opacities. Mice received 0.5, 1 and 2 Gy 60Co gamma-ray irradiation at dose rates of 0.063 and 0.3 Gy min-1. Scheimpflug imaging was used to quantify lens opacification as an early indicator of cataract, with monthly observations taken postirradiation for an 18-month period in all strains apart from 129S2, which were observed for 12 months. Opacification of the lens was found to increase with time postirradiation (with age) for most mouse models, with ionizing radiation exposure increasing opacities further. Sex, dose, dose rate and genetic background were all found to be significant contributors to opacification; however, significant interactions were identified, which meant that the impact of these factors was strain dependent. Mean lens density increased with higher dose and dose rate in the presence of Ercc2 and Ptch1 mutations. This project was the first to focus on low (<1 Gy) dose, multiple dose rate, sex and strain effects in lens opacification, and clearly demonstrates the importance of these experimental factors in radiobiological investigations on the lens. The results provide insight into the effects of ionizing radiation on the lens as well as the need for further work in this area to underpin appropriate radiation protection legislation and guidance.


Asunto(s)
Catarata/etiología , Animales , Femenino , Antecedentes Genéticos , Humanos , Cristalino/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Exposición Profesional , Receptor Patched-1/metabolismo , Dosis de Radiación , Exposición a la Radiación , Radiación Ionizante , Proteína de la Xerodermia Pigmentosa del Grupo D/metabolismo
8.
DNA Repair (Amst) ; 74: 70-79, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30606609

RESUMEN

DSBs are harmful lesions produced through endogenous metabolism or by exogenous agents such as ionizing radiation, that can trigger genomic rearrangements. We have recently shown that exposure to 2 Gy of X-rays has opposite effects on the induction of Shh-dependent MB in NHEJ- and HR-deficient Ptch1+/- mice. In the current study we provide a comprehensive link on the role of HR/NHEJ at low doses (0.042 and 0.25 Gy) from the early molecular changes through DNA damage processing, up to the late consequences of their inactivation on tumorigenesis. Our data indicate a prominent role for HR in genome stability, by preventing spontaneous and radiation-induced oncogenic damage in neural precursors of the cerebellum, the cell of origin of MB. Instead, loss of DNA-PKcs function increased DSBs and apoptosis in neural precursors of the developing cerebellum, leading to killing of tumor initiating cells, and suppression of MB tumorigenesis in DNA-PKcs-/-/Ptch1+/- mice. Pathway analysis demonstrates that DNA-PKcs genetic inactivation confers a remarkable radiation hypersensitivity, as even extremely low radiation doses may deregulate many DDR genes, also triggering p53 pathway activation and cell cycle arrest. Finally, by showing that DNA-PKcs inhibition by NU7441 radiosensitizes human MB cells, our in vitro findings suggest the inclusion of MB in the list of tumors beneficiating from the combination of radiotherapy and DNA-PKcs targeting, holding promise for clinical translation.


Asunto(s)
Neoplasias Cerebelosas/genética , Reparación del ADN/efectos de la radiación , Meduloblastoma/genética , Neoplasias Inducidas por Radiación/genética , Receptor Patched-1/deficiencia , Receptor Patched-1/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/efectos de la radiación , Línea Celular Tumoral , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Neoplasias Cerebelosas/terapia , Daño del ADN , Reparación del ADN por Unión de Extremidades/efectos de la radiación , ADN Helicasas/genética , Proteína Quinasa Activada por ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Relación Dosis-Respuesta en la Radiación , Recombinación Homóloga/efectos de la radiación , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Meduloblastoma/terapia , Ratones , Terapia Molecular Dirigida , Mutación , Neoplasias Inducidas por Radiación/metabolismo , Neoplasias Inducidas por Radiación/patología , Neoplasias Inducidas por Radiación/terapia , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Riesgo , Rayos X/efectos adversos
9.
Radiat Environ Biophys ; 57(1): 5-15, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29247291

RESUMEN

MELODI (Multidisciplinary European Low Dose Initiative) is a European radiation protection research platform with focus on research on health risks after exposure to low-dose ionising radiation. It was founded in 2010 and currently includes 44 members from 18 countries. A major activity of MELODI is the continuous development of a long-term European Strategic Research Agenda (SRA) on low-dose risk for radiation protection. The SRA is intended to identify priorities for national and European radiation protection research programs as a basis for the preparation of competitive calls at the European level. Among those key priorities is the improvement of health risk estimates for exposures close to the dose limits for workers and to reference levels for the population in emergency situations. Another activity of MELODI is to ensure the availability of European key infrastructures for research activities, and the long-term maintenance of competences in radiation research via an integrated European approach for training and education. The MELODI SRA identifies three key research topics in low dose or low dose-rate radiation risk research: (1) dose and dose rate dependence of cancer risk, (2) radiation-induced non-cancer effects and (3) individual radiation sensitivity. The research required to improve the evidence base for each of the three key topics relates to three research lines: (1) research to improve understanding of the mechanisms contributing to radiogenic diseases, (2) epidemiological research to improve health risk evaluation of radiation exposure and (3) research to address the effects and risks associated with internal exposures, differing radiation qualities and inhomogeneous exposures. The full SRA and associated documents can be downloaded from the MELODI website ( http://www.melodi-online.eu/sra.html ).


Asunto(s)
Comunicación Interdisciplinaria , Dosis de Radiación , Radiobiología/métodos , Humanos , Exposición a la Radiación , Tolerancia a Radiación , Medición de Riesgo
10.
Radiat Res ; 179(5): 610-4, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23560628

RESUMEN

Medulloblastomas in Patched heterozygous mice (Ptc1(+/-) mice) are induced with high probability by ionizing radiation applied in the immediate post-natal period. A mathematical model is described here that accommodates the dependence of the medulloblastoma incidence on dose, age at exposure and age. The model assumes that the first step in the development of the cancer is already present in all cells of the patched mouse due to germ-line inactivation of one allele of the patched tumor suppressor gene. The subsequent rate-limiting step is dependent linearly on dose at least up to 3 Gy. The observed strong decrease in carcinogenic effect of radiation between exposure on day 1 and day 10 is described by a physiological elimination of target cells during post-natal maturation of the brain. A single malignant cell develops into a tumor following a gamma-distribution with mean of about 160 days. The multiplicity of medulloblastomas is predicted.


Asunto(s)
Neoplasias Cerebelosas/patología , Meduloblastoma/patología , Modelos Biológicos , Neoplasias Inducidas por Radiación/patología , Animales , Neoplasias Cerebelosas/genética , Progresión de la Enfermedad , Relación Dosis-Respuesta en la Radiación , Heterocigoto , Estimación de Kaplan-Meier , Funciones de Verosimilitud , Meduloblastoma/genética , Ratones , Mutación , Neoplasias Inducidas por Radiación/genética , Distribución de Poisson , Factores de Tiempo
11.
Curr Mol Med ; 12(5): 613-24, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22452594

RESUMEN

A long-held dogma in radiation biology has been that the biological effects of exposure to ionizing radiation occur as a result of damage in directly irradiated cells and that no effect would occur in neighboring unirradiated cells. This paradigm has been frequently challenged by reports of radiation effects in unirradiated or 'bystander' cells receiving signals from directly irradiated cells, an issue that may have substantial impact on radiation risk assessment and development of radiation-based therapies. Radiation-induced bystander effects have been shown in single-cell systems in vitro for an array of cancer relevant endpoints, and may trigger damage in more complex 3-D tissue systems. They may be mediated by soluble factors released by irradiated cells into the extracellular environment and/or by the passage of mediator molecules through gap-junction intercellular communication. To date, evidence that radiation-associated bystander or abscopal responses are effectual in vivo has been limited, but new data suggest that they may significantly affect tumor development in susceptible mouse models. Further understanding of how the signal/s is transmitted to unirradiated cells and tissues and how it provokes long-range and significant responses is crucial. By summarizing the existing evidence of radiation induced bystander-like effects in various systems with emphasis on in vivo findings, we will discuss the potential mechanisms involved in these observations and how effects in bystander cells contribute to uncertainties in assessing cancer risks associated with radiation exposure.


Asunto(s)
Efecto Espectador/fisiología , Radiación Ionizante , Animales , Comunicación Celular , Daño del ADN/efectos de la radiación , Humanos , Neoplasias/etiología
12.
Oncogene ; 30(45): 4601-8, 2011 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-21602884

RESUMEN

Ionizing radiation is a genotoxic agent and human carcinogen. Recent work has questioned long-held dogmas by showing that cancer-associated genetic alterations occur in cells and tissues not directly exposed to radiation, questioning the robustness of the current system of radiation risk assessment. In vitro, diverse mechanisms involving secreted soluble factors, gap junction intercellular communication (GJIC) and oxidative metabolism are proposed to mediate these indirect effects. In vivo, the mechanisms behind long-range 'bystander' responses remain largely unknown. Here, we investigate the role of GJIC in propagating radiation stress signals in vivo, and in mediating radiation-associated bystander tumorigenesis in mouse central nervous system using a mouse model in which intercellular communication is downregulated by targeted deletion of the connexin43 (Cx43) gene. We show that GJIC is critical for transmission of oncogenic radiation damage to the non-targeted cerebellum, and that a mechanism involving adenosine triphosphate release and upregulation of Cx43, the major GJIC constituent, regulates transduction of oncogenic damage to unirradiated tissues in vivo. Our data provide a novel hypothesis for transduction of distant bystander effects and suggest that the highly branched nervous system, similar to the vascular network, has an important role.


Asunto(s)
Adenosina Trifosfato/metabolismo , Efecto Espectador/efectos de la radiación , Transformación Celular Neoplásica/genética , Neoplasias Cerebelosas/genética , Conexina 43/metabolismo , Daño del ADN/genética , Neoplasias Inducidas por Radiación/genética , Animales , Cerebelo/metabolismo , Cerebelo/efectos de la radiación , Conexina 43/genética , Uniones Comunicantes/metabolismo , Uniones Comunicantes/efectos de la radiación , Ratones , Dosis de Radiación , Eliminación de Secuencia/efectos de la radiación , Transducción de Señal/efectos de la radiación
13.
Oncogene ; 30(47): 4740-9, 2011 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-21602895

RESUMEN

Heterozygous Patched1 (Ptc1(+/-)) mice are prone to medulloblastoma (MB), and exposure of newborn mice to ionizing radiation dramatically increases the frequency and shortens the latency of MB. In Ptc1(+/-) mice, MB is characterized by loss of the normal remaining Ptc1 allele, suggesting that genome rearrangements may be key events in MB development. Recent evidence indicates that brain tumors may be linked to defects in DNA-damage repair processes, as various combinations of targeted deletions in genes controlling cell-cycle checkpoints, apoptosis and DNA repair result in MB in mice. Non-homologous end joining (NHEJ) and homologous recombination (HR) contribute to genome stability, and deficiencies in either pathway predispose to genome rearrangements. To test the role of defective HR or NHEJ in tumorigenesis, control and irradiated Ptc1(+/-) mice with two, one or no functional Rad54 or DNA-protein kinase catalytic subunit (DNA-PKcs) alleles were monitored for MB development. We also examined the effect of Rad54 or DNA-PKcs deletion on the processing of endogenous and radiation-induced double-strand breaks (DSBs) in neural precursors of the developing cerebellum, the cells of origin of MB. We found that, although HR and NHEJ collaborate in protecting cells from DNA damage and apoptosis, they have opposite roles in MB tumorigenesis. In fact, although Rad54 deficiency increased both spontaneous and radiation-induced MB development, DNA-PKcs disruption suppressed MB tumorigenesis. Together, our data provide the first evidence that Rad54-mediated HR in vivo is important for suppressing tumorigenesis by maintaining genomic stability.


Asunto(s)
Neoplasias Cerebelosas/etiología , Reparación del ADN por Unión de Extremidades , Recombinación Homóloga , Meduloblastoma/etiología , Receptores de Superficie Celular/fisiología , Animales , Neoplasias Cerebelosas/genética , Daño del ADN , ADN Helicasas/fisiología , Proteína Quinasa Activada por ADN/fisiología , Inestabilidad Genómica , Pérdida de Heterocigocidad , Meduloblastoma/genética , Ratones , Proteínas Nucleares/fisiología , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Riesgo
14.
Carcinogenesis ; 30(2): 340-7, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18952596

RESUMEN

Patched1 heterozygous mice (Ptch1(+/-)) are useful for basal cell carcinoma (BCC) studies, being remarkably susceptible to BCC induction by ultraviolet or ionizing radiation. Analogously, skin carcinogenesis-susceptible (Car-S) mice are elective for studies of papilloma and squamous cell carcinoma (SCC) induction. We previously reported a striking effect of gender on BCC induction in Ptch1(+/-) mice, with total resistance of females; likewise, Car-S females show increased skin tumor resistance relative to males. Here, we investigated the protective role of endogenous estrogen in skin keratinocyte tumorigenesis. Control (CN) and ovariectomized Ptch1(+/-) or Car-S females were irradiated for BCC induction or topically treated with chemical carcinogens for SCC induction. Susceptibility to BCC or SCC was dramatically increased in ovariectomized Ptch1(+/-) and Car-S females and restored to levels observed in males. Remarkably, progression of initially benign papillomas to malignant SCC occurred only in ovariectomized Car-S females. We explored the mechanisms underlying tumor progression and report overexpression of estrogen receptor (ER)-alpha, downregulation of ERbeta and upregulation of cyclin D1 in papillomas from ovariectomized Car-S relative to papillomas from CN females. Thus, an imbalanced ERalpha/ERbeta expression may be associated with estrogen-mediated modulation of non-melanoma skin carcinogenesis, with a key role played by cyclin D1. Our findings underscore a highly protective role of endogenous estrogen against skin tumorigenesis by diverse agents in two independent mouse models of skin cancer.


Asunto(s)
Carcinoma Basocelular/metabolismo , Carcinoma de Células Escamosas/metabolismo , Estrógenos/fisiología , Neoplasias Cutáneas/metabolismo , Animales , Carcinoma Basocelular/patología , Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclina D1/metabolismo , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Masculino , Ratones , Neoplasias Inducidas por Radiación/metabolismo , Neoplasias Inducidas por Radiación/patología , Ovariectomía , Papiloma/metabolismo , Papiloma/patología , Receptores Patched , Receptor Patched-1 , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Neoplasias Cutáneas/patología , Rayos Ultravioleta
15.
Homo ; 59(6): 439-52, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18995852

RESUMEN

The objective of the present study was to investigate the psycho-social health and weight status of two samples of North African immigrants measured in 1990 (166 males) and in 2000-2002 (173 males and females), respectively. In addition to the cross-sectional study, we conducted a repeated study on a sub-sample of 21 males measured both in 1990 and in 2000-2002. The study was carried out in Italian health and care dedicated centres spread all over the Bologna administrative areas, that belong to the AUSL (Azienda Unità Sanitaria Locale, Administrative Local Health Unit). To evaluate the health and weight status, we calculated the body mass index (BMI) and measured systolic and diastolic blood pressure. Questionnaires were used to assess the psycho-social status. The mean BMI increased significantly (p<0.05) after immigration, as the prevalence of overweight (BMI>25) was higher after a decade. There was a significant increase (p<0.001) in stress-related factors and in the desire to return home. We also recorded an increase in weight disorders, as the length of time since immigration was an important risk factor for overweight. Therefore, immigrants may have a high risk of obesity-related co-morbidities. However, the factors related to malnutrition and stress had only weak effects on blood pressure.


Asunto(s)
Emigrantes e Inmigrantes/psicología , Psicología , Adulto , África del Norte/etnología , Índice de Masa Corporal , Peso Corporal , Etnicidad , Femenino , Estado de Salud , Humanos , Italia , Masculino , Marruecos/etnología , Trastornos Nutricionales/epidemiología , Obesidad/epidemiología , Sobrepeso/epidemiología , Sístole
16.
Radiat Res ; 168(6): 733-40, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18088186

RESUMEN

Patched1 heterozygous knockout mice (Ptc1+/-), an animal model of multiorgan tumorigenesis in which ionizing radiation dramatically accelerates tumor development, were used to study the potential tumorigenic effects of electromagnetic fields (EMFs) on neonatal mice. Two hundred Ptc1+/- mice and their wild-type siblings were enrolled in this study. Newborn mice were exposed to 900 MHz radiofrequency radiation (average SAR: 0.4 W/kg for 5 days, 0.5 h twice a day) or were sham exposed. We found that RF EMFs simulating the Global System for Mobile Communications (GSM) did not affect the survival of the mice, because no statistically significant differences in survival were found between exposed and sham-exposed animals. Also, no effects attributable to radiofrequency radiation were observed on the incidence and histology of Ptc1-associated cerebellar tumors. Moreover, the skin phenotype was analyzed to look for proliferative effects of RF EMFs on the epidermal basal layer and for acceleration of preneoplastic lesions typical of the basal cell carcinoma phenotype of this model. We found no evidence of proliferative or promotional effects in the skin from neonatal exposure to radiofrequency radiation. Furthermore, no difference in Ptc1-associated rhabdomyosarcomas was detected between sham-exposed and exposed mice. Thus, under the experimental conditions tested, there was no evidence of life shortening or tumorigenic effects of neonatal exposure to GSM RF radiation in a highly tumor-susceptible mouse model.


Asunto(s)
Heterocigoto , Ondas de Radio , Receptores de Superficie Celular/metabolismo , Animales , Animales Recién Nacidos , Supervivencia sin Enfermedad , Campos Electromagnéticos , Ratones , Ratones Transgénicos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/radioterapia , Receptores Patched , Receptor Patched-1 , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Lesiones Precancerosas/radioterapia , Receptores de Superficie Celular/genética , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/radioterapia
17.
Oncogene ; 25(55): 7267-73, 2006 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-16878160

RESUMEN

Medulloblastoma (MB) results from aberrant development of cerebellar neurons in which altered hedgehog (Hh) signalling plays a major role. We investigated the possible influence of Hh signalling on ErbB-receptor expression in MB, in particular that of the ErbB-4 CYT-1 and CYT-2 isoforms generated by alternative splicing of the cytoplasmic domain. ErbB-4 expression was downregulated in Hh-induced MBs from Patched-1(+/-) mice. Hh signalling (reflected by enhanced expression of the Gli1 transcription factor) inhibited ErbB-4 expression in mouse cerebellar granule progenitors and human MB cells. Analysis of 26 human primary MBs revealed a subset of 11 tumors characterized by low Gli1 levels, upregulated ErbB-4 expression and increased CYT-1:CYT-2 ratios. Interestingly, CYT-1 and Gli1 levels were inversely correlated. ErbB-4 CYT-1 and CYT-2 had different phenotypic effects in cultured MB cells: in response to neuregulin treatment, CYT-2 overexpression inhibited proliferation whereas CYT-1, which includes a phosphatidylinositol 3-kinase (PI3K)-binding site that is missing in CYT-2, enhanced resistance to starvation- and etoposide-induced apoptosis by activating PI3K/Akt signalling. CYT-1:CYT-2 ratios displayed correlation with tumor histotype and ErbB-2 levels, which are established prognostic indices for MB. These findings demonstrate that low-level Hh signalling in human MB is associated with the selective maintenance of high ErbB-4 CYT-1 expression, an alteration that exerts tumor-promoting effects.


Asunto(s)
Empalme Alternativo , Citoplasma/metabolismo , Receptores ErbB/metabolismo , Proteínas Hedgehog/metabolismo , Meduloblastoma/clasificación , Transducción de Señal , Animales , Secuencia de Bases , Cartilla de ADN , Humanos , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones , Pronóstico , Receptor ErbB-4 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Oncogene ; 25(40): 5575-80, 2006 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-16636673

RESUMEN

Inactivation of one Ptc1 allele predisposes humans and mice to spontaneous medulloblastoma development, and irradiation of newborn Ptc1 heterozygous mice results in dramatic increase of medulloblastoma incidence. While a role for loss of wild-type (wt) Ptc1 (LOH) in radiation-induced medulloblastomas from Ptc1(neo67/+) mice is well established, the importance of this event in spontaneous medulloblastomas is still unclear. Here, we demonstrate that biallelic Ptc1 loss plays a crucial role in spontaneous medulloblastomas, as shown by high rate of wt Ptc1 loss in spontaneous tumors. In addition, remarkable differences in chromosomal events involving the Ptc1 locus in spontaneous and radiation-induced medulloblastomas suggest distinct mechanisms for Ptc1 loss. To assess when, during tumorigenesis, Ptc1 loss occurs, we characterized cerebellar abnormalities that precede tumor appearance in Ptc1(neo67/+) mice. We show that inactivation of only one copy of Ptc1 is sufficient to give rise to abnormal cerebellar proliferations with different degree of altered cell morphology, but lacking potential to progress to neoplasia. Furthermore, we identify biallelic Ptc1 loss as the event causally related to the transition from the preneoplastic stage to full blown medulloblastoma. These results underscore the utility of the Ptc1(neo67/+) mouse model for studies on the mechanisms of medulloblastoma and for development of new therapeutic strategies.


Asunto(s)
Neoplasias Cerebelosas/genética , Meduloblastoma/genética , Lesiones Precancerosas/genética , Receptores de Superficie Celular/genética , Envejecimiento , Animales , Neoplasias Cerebelosas/patología , Cromosomas de los Mamíferos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Proteínas Hedgehog , Pérdida de Heterocigocidad , Meduloblastoma/patología , Ratones , Ratones Endogámicos C57BL , Receptores Patched , Receptor Patched-1 , Lesiones Precancerosas/patología , Radiación Ionizante , Transducción de Señal , Transactivadores/metabolismo
19.
Oncogene ; 25(8): 1165-73, 2006 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-16407852

RESUMEN

Hemizygous Ptc1 mice have many features of Gorlin syndrome, including predisposition to medulloblastoma development. Ionizing radiation synergize with Ptc1 mutation to induce medulloblastoma only in neonatally exposed mice. To explore the mechanisms underlying age-dependent susceptibility, we irradiated Ptc(neo67/+) mice at postnatal day 1 (P1) or 10 (P10). We observed a dramatic difference in medulloblastoma incidence, which ranged from 81% in the cerebellum irradiated at P1 to 3% in the cerebellum irradiated at P10. A striking difference was also detected in the frequency of cerebellar preneoplastic lesions (100 versus 14%). Our data also show significantly lower induction of apoptosis in the cerebellum of medulloblastoma-susceptible (P1) compared to -resistant (P10) mice, strongly suggesting that medulloblastoma formation in Ptc1 mutants may be associated with resistance to radiation-induced cell killing. Furthermore, in marked contrast with P10 mice, cerebellum at P1 displays substantially increased activation of the cell survival-promoting Akt/Pkb protein, and markedly decreased p53 levels in response to radiation-induced genotoxic stress. Overall, these results show that developing cerebellar granule neuron precursors' (CGNPs) radiosensitivity to radiation-induced cell death increases with progressing development and inversely correlates with their ability to neoplastically transform.


Asunto(s)
Transformación Celular Neoplásica/efectos de la radiación , Neoplasias Cerebelosas/etiología , Daño del ADN/efectos de la radiación , Heterocigoto , Meduloblastoma/etiología , Neoplasias Inducidas por Radiación/genética , Desequilibrio Alélico , Animales , Animales Recién Nacidos , Apoptosis/efectos de la radiación , Neoplasias Cerebelosas/patología , Cerebelo/efectos de la radiación , Incidencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Meduloblastoma/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Superficie Celular , Proteína p53 Supresora de Tumor/metabolismo , Rayos X
20.
Mamm Genome ; 12(4): 291-4, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11309660

RESUMEN

Car-R and Car-S outbred mouse lines, phenotypically selected for resistance and susceptibility to skin carcinogenesis respectively, show significant linkage disequilibrium (LD) at genetic markers mapping on chromosomal regions where skin cancer modifier loci (Skts3, Skts1, and Psl1 on Chrs 5, 7, and 9 respectively) have been mapped in standard crosses. Analysis of these regions for genetic linkage with skin cancer phenotypes in 245 (Car-R x Car-S)F2 intercross mice, by using single nucleotide polymorphisms (SNPs), revealed significant linkage at a possible allelic form of the Skts1 locus, whose mapping region was shortened to a <5.5-cM interval near the Tyr locus. The Car-derived Skts1 locus was linked with papilloma multiplicity and latency by a recessive inheritance of the susceptibility allele. Putative loci on Chr 5 (Skts3) and 9 (Psl1) showed no significant linkage. These results point to the important role of the Stks1 locus in mouse skin tumorigenesis in independent crosses. The shortened Skts1 mapping region should facilitate the identification of candidate genes.


Asunto(s)
Desequilibrio de Ligamiento , Polimorfismo de Nucleótido Simple/genética , Neoplasias Cutáneas/genética , Animales , Animales no Consanguíneos , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Haplotipos/genética , Heterocigoto , Ratones , Ratones Endogámicos , Repeticiones de Microsatélite
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...