Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
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.
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
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.
Oncotarget ; 8(55): 94247-94258, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29212225

ABSTRACT

Along with their role in the maintenance of nuclear architecture, nuclear lamins also control genomic stability, DNA damage repair, transcription, cell proliferation, differentiation and senescence. Recent reports reveal that prelamin A-processing defects play a role in cancer development by impacting on transcription of key players in the maintenance of the genome stability, including RAD51. Here, we performed a 'proof of concept' study evaluating the role of prelamin A and RAD51 expression in clinical outcome of cervical cancer patients. We analyzed biomarker expression by immunohistochemistry in tumor material from locally advanced cervical cancer (LACC) patients (n=66) and correlated data with clinicopathological parameters and with response to neoadjuvant chemoradiation (CT/RT). In LACC patients who underwent neoadjuvant CT/RT the percentage of cases showing high prelamin A levels was greater in patients who completely responded to treatment (25 of 40, 62.5%) than in patients with macroscopic residual tumor (6 of 26, 23.1%, p=0.0024). Conversely, patients showing high RAD51 expression were less likely to respond to treatment (14 of 26, 53.8%) than were those with low protein levels (12 of 40, 30%, p=0.072). Only prelamin A retained an independent role in predicting response to treatment (p=0.003), while RAD51 approached statistical significance (p=0.07). Notably, high RAD51 expression highly significantly predicted poor outcome, emerging as an independent prognostic factor for disease free survival (p=0.038), while approaching statistical significance for overall survival (p=0.09). Our findings provide a framework for future prospective studies investigating molecular predictors of response to neoadjuvant chemoradiotherapy in LACC patients.

5.
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.

6.
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
7.
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
8.
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
9.
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
10.
PLoS One ; 9(7): e101623, 2014.
Article in English | MEDLINE | ID: mdl-25000562

ABSTRACT

BACKGROUND: The male-to-female sex ratio for medulloblastoma (MB) is approximately 1.5∶1, female gender being also a favorable prognostic factor. This study aimed at evaluating the impact of gender on MB tumorigenesis. METHODS: In vitro activity of 17ß-estradiol (E2), DPN [2,3-bis(4-hydroxyphenyl)-propionitrile, a selective estrogen receptor ß (ERß)-agonist], PPT [4,4',4″-(4-Propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol, a selective ERα-agonist] or DHT (5 alpha-dihydrotestosterone) was evaluated in three human MB cell lines. D283 Med cells were transplanted into athymic mice. RESULTS: A significant expression of ERß, with little or no ERα, and low AR (androgen receptor) was found in MB cell lines. The compounds tested did not affect cell proliferation. In vivo, we observed a significantly lower growth of D283 Med in nude female mice compared to males. At microscopic examination, tumors from females showed a shift towards differentiation, as evaluated by lower nestin, and higher NSE (neuron-specific enolase) and GFAP (glial fibrillary acidic protein) expression compared to males. Tumors from females also showed lower Ki67 and p53 expression. The wild-type ERß protein (ERß1) was lost in male tumors, while it was a permanent feature in females, and a strong negative correlation was found between Ki67 and ERß1 expression. Conversely, tumor levels of ERß2 and ERß5 did not significantly differ between genders. Increased levels of cyclin-dependent kinase inhibitor p21 were observed in females, suggesting that estrogen may decrease tumor growth through blocking cell cycle progression. An inhibition of the insulin-like growth factor I (IGF-I) signaling was also evident in females. CONCLUSION: We provides mechanistic evidence supporting the idea that ERß1 signaling may have pro-differentiation and tumor suppressive function in medulloblastomas.


Subject(s)
Carcinogenesis/metabolism , Cerebellar Neoplasms/pathology , Estrogen Receptor beta/metabolism , Medulloblastoma/pathology , Sex Characteristics , Signal Transduction , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/physiopathology , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estrogen Receptor beta/agonists , Female , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/physiopathology , Mice , Nitriles/pharmacology , Phenols/pharmacology , Pyrazoles/pharmacology , Signal Transduction/drug effects
11.
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
12.
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
13.
PLoS One ; 8(3): e60145, 2013.
Article in English | MEDLINE | ID: mdl-23555905

ABSTRACT

Recent evidence links aberrant activation of Hedgehog (Hh) signaling with the pathogenesis of several cancers including medulloblastoma, glioblastoma, melanoma as well as pancreas, colorectal, and prostate carcinomas. Here we investigated the role of the transcription factor Gli1 in ovarian cancer. To this end, the expression profile of Gli1 was examined in normal ovaries, ovarian tumors, and ovarian cancer cell lines, and the in vitro effects of a specific Hh-pathway blocker, KAAD-cyclopamine, or a specific Gli1 inhibitor (GANT58) on cell proliferation and on Hh target gene expression were also assessed. Results obtained showed that epithelial cells in ovarian cancer tissue express significantly higher levels of nuclear Gli1 than in normal ovarian tissue, where the protein was almost undetectable. In addition, multivariate analysis showed that nuclear Gli1 was independently associated to poor survival in advanced serous ovarian cancer patients (HR = 2.2, 95%CI 1.0-5.1, p = 0.04). In vitro experiments demonstrated Gli1 expression in the three ovarian carcinoma cell lines tested, A2780, SKOV-3 and OVCAR-3. Remarkably, although KAAD-cyclopamine led to decreased cell proliferation, this treatment did not inhibit hedgehog target gene expression in any of the three ovarian cancer cell lines, suggesting that the inhibition of cell proliferation was a nonspecific or toxic effect. In line with these data, no differences on cell proliferation were observed when cell lines were treated with GANT58. Overall, our clinical data support the role of Gli1 as a prognostic marker in advanced serous ovarian cancer and as a possible therapeutic target in this disease. However, our in vitro findings draw attention to the need for selection of appropriate experimental models that accurately represent human tumor for testing future therapies involving Hh pathway inhibitors.


Subject(s)
Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Transcription Factors/metabolism , Adult , Aged , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cinnamates/pharmacology , Female , Humans , Immunoprecipitation , Middle Aged , Ovarian Neoplasms/genetics , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1
14.
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
15.
Hum Pathol ; 44(6): 1047-54, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23266443

ABSTRACT

The prognostic relevance of estrogen (ER) and progesterone receptor (PR) expression in endometrioid endometrial cancer is still controversially discussed. The present study has focused on the evaluation of the prognostic value of ERα, ERß1, ERß2, and PR in this histotype. Specifically, we were interested in evaluating whether the relative level of ER subtype-specific expression (in terms of a ratio ERα/ERß1 and ERα/ERß2) would predict clinical outcome better than their absolute levels in patients with endometrioid endometrial cancer. To this end, protein content was assessed by immunohistochemistry in a group of 121 cases and staining was analyzed in relation to clinicopathologic variables, disease-free survival and overall survival. Results obtained have demonstrated that none of the biological markers analyzed possess an independent prognostic role with regard to disease-free survival. Multivariate analysis of overall survival has shown that ERα alone is not an independent prognostic indicator in patients with endometrioid endometrial cancer (hazard ratio [HR]; 0.5; 95% confidence interval [CI], 0.09-3.0; P = .5). On the other hand, an ERα/ERß1 ratio of 1 or less or an ERα/ERß2 ratio of 1 or less has proved to be independently associated with a higher risk of death (HR, 6.4 [95% CI, 1.0-40.6; P = .04] and 9.7 [95% CI, 1.1-85.3; P = .04], respectively) along with age, tumor stage, and Ki-67. In conclusion, we report here that the ERα/ERß1 and ERα/ERß2 expression ratios are independent prognostic markers of survival in endometrioid endometrial cancer; these findings suggest that phenotyping these interacting markers conjointly may better predict patient survival than each individual marker alone.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor beta/biosynthesis , Adult , Aged , Aged, 80 and over , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Disease-Free Survival , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Estrogen Receptor alpha/analysis , Estrogen Receptor beta/analysis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis
16.
Int J Gynecol Pathol ; 31(6): 507-16, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23018221

ABSTRACT

Two types of ovarian carcinomas are distinguished with respect to morphology, biology, and clinical course, and are designated as Type I and Type II tumors. However, placement of clear cell carcinomas into one of these 2 groups has been problematic as they exhibit morphologic, molecular, and clinical features that do not entirely resemble either Type I or Type II tumors. The present study aimed at better elucidating the clinicopathologic and immunohistochemical features of clear cell carcinomas, in comparison with the 2 main broad categories. To this end, a panel of classic clinicopathologic and immunohistochemical parameters, including estrogen receptor α (ERα), ERß, progesterone receptor, Ki67, p53, and HER2/neu was evaluated in 71 Type I, 157 Type II, and 21 clear cell carcinomas. Overall, findings from the present study support the idea that ovarian clear cell carcinomas are neither Type I nor Type II carcinomas of the ovary; indeed, results obtained showed that similarities between clear cell carcinomas and Type I were limited to the patient's age, tumor dimension, incidence of lymph node and extranodal metastases, and p53 labeling index, whereas the patient's age and incidence of extranodal metastases were the only parameters comparable with the Type II group. The hormonal receptor profile of clear cell carcinomas was characterized by low expression of nuclear ERα and progesterone receptor, and by almost exclusively nuclear ERß immunopositivity, features significantly different from both Type I and II tumors. Finally, the percentage of HER2/neu-positive samples in clear cell carcinomas was 10- and 2.5-fold higher than Type I and Type II ovarian tumors, respectively. In conclusion, our study provides insights into clear cell carcinoma that could help in explaining its unique prognostic features, and, eventually, in orienting toward new therapeutic options.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/chemistry , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/chemistry , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
17.
Curr Pharm Des ; 18(19): 2734-57, 2012.
Article in English | MEDLINE | ID: mdl-22390760

ABSTRACT

While it is well documented that the mitogenic actions of estrogens are critical in the development and progression of human breast and some gynecologic cancers, only latest data demonstrate a crucial involvement of estrogen-signaling in the carcinogenesis of non-classical estrogen target tissues, as colon, prostate, lung, skin, and brain. Only recently it has also been found out that the biological effects of estrogens are mediated by two distinct estrogen receptors (ERs), ERα and ERß, and that their relative levels in a given cell are important determinants of response to estradiol and selective estrogen receptor modulators. Indeed, although ERα and ERß have similar structure, they produce different effects, and there is currently increasing evidence that, for some tumors, an imbalanced ERß expression might play a pivotal role in tumor development and progression. However, the prognostic value, the potential significance in predicting response to endocrine therapy, and, eventually, the utility of ERß as a therapeutic target need to be assessed in large-scale and prospective clinical studies. This review examines the experimental and clinical evidences for a role of ERß in carcinogenesis of classical and nonclassical estrogen target tissues. If anomalies of ERß expression could be demonstrated to represent a critical step in the development and progression of some types of cancers, its re-expression by genetic engineering, as well as the use of targeted ERß therapies would constitute new important therapeutic approaches.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Delivery Systems , Estrogen Receptor beta/drug effects , Breast Neoplasms/metabolism , Estrogen Receptor beta/metabolism , Female , Humans , Signal Transduction
18.
Hum Pathol ; 43(2): 282-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21835429

ABSTRACT

In this study, we investigated the prognostic value of metastasis tumor antigen 1 expression in 81 untreated patients with ovarian cancer. The expression of metastasis tumor antigen 1 was evaluated by immunohistochemistry, and staining was analyzed in relation to clinicopathologic variables, disease-free survival, and overall survival. High expression of metastasis tumor antigen 1 was found to be associated with advanced stage (I/II versus III/IV, P = .02) and with worse response to first-line treatment (P = .03). Cases with high metastasis tumor antigen 1 expression showed a lower disease-free survival compared with cases with low expression (P = .02). In multivariate analysis of disease-free survival, metastasis tumor antigen 1 overexpression retained an independent negative prognostic role (P = .04), when considered together with histotype, stage of disease, residual tumor at surgery, and chemosensitivity. The evaluation of the prognostic relevance of metastasis tumor antigen 1 in late-stage disease showed that overexpression was a prognostic factor for poor disease-free survival and overall survival in this subset of patients, in both univariate and multivariate models. These findings indicate that metastasis tumor antigen 1 overexpression can be used as a predictor of clinical outcome in patients with ovarian cancer and therefore may represent a new prognostic marker.


Subject(s)
Adenocarcinoma/secondary , Histone Deacetylases/metabolism , Ovarian Neoplasms/pathology , Repressor Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Survival Analysis , Survival Rate , Taiwan/epidemiology , Trans-Activators
19.
Histopathology ; 59(5): 909-17, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22092402

ABSTRACT

AIMS: To investigate the prognostic value of cytoplasmic oestrogen receptor beta (ERß) expression in a series of untreated patients with non-human papillomavirus (HPV)-related vulvar cancer. METHODS AND RESULTS: Immunohistochemistry was carried out using a polyclonal rabbit anti-human ERß antibody. The nuclear and cytoplasmic expression of ERß was evaluated in 33 patients. Cytoplasmic immunoreactivity was correlated with histopathological and molecular parameters (Ki67, p21), disease-free survival (DFS) and overall survival (OS). The expression of cytoplasmic ERß was found to be associated with grade (P=0.006), while no association was found with any of the remaining variables examined. Cases with high cytoplasmic ERß expression showed lower DFS and OS compared to cases with low cytoplasmic ERß (P=0.007, P=0.01, respectively). There was also a progressive decline in both the DFS and OS with increasing tumour size (P=0.05, P=0.07, respectively) and with increasing depth of infiltration (P=0.14, P=0.07, respectively). On multivariate analysis, only tumour size and cytoplasmic ERß staining retained an independent negative prognostic role for DFS and OS. CONCLUSIONS: The assessment of cytoplasmic ERß expression could be helpful to identify poor prognosis in elderly patients with non-HPV-related vulvar squamous cell carcinoma (SCC).


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cytoplasm/metabolism , Estrogen Receptor beta/biosynthesis , Vulvar Neoplasms/metabolism , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Estrogen Receptor beta/analysis , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Vulvar Neoplasms/mortality , Vulvar Neoplasms/pathology
20.
Gynecol Oncol ; 122(3): 573-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21665249

ABSTRACT

OBJECTIVE: In this study we investigated the prognostic value of estrogen receptor α (ERα), ERß and progesterone receptor (PR) expression in 58 untreated advanced serous ovarian cancer patients. The study also included 12 macroscopically and histopathologically normal ovaries. MATERIALS AND METHODS: Protein expression was evaluated by immunohistochemistry, and antibody staining detected in both the nuclear and cytoplasmic compartments was taken into account. Immunopositivity was analyzed in relation to tumor clinicopathological variables, disease-free survival (DFS), and overall survival (OS). RESULTS: Epithelial cells in ovarian cancer tissue showed significantly lower levels of nuclear ERß and PR, but not ERα, than in normal ovarian tissue. In the case of ERß, however, while normal ovarian epithelium exhibited almost exclusively strong nuclear staining, ovarian cancer tissue mostly showed cytoplasmic immunopositivity. Nuclear ERα and ERß expression were not associated with clinical outcome. Conversely, any cytoplasmic ERß expression was an independent unfavorable prognostic factor for DFS, a finding approaching statistical significance also for OS. These data suggest that, in advanced serous ovarian cancer, cytoplasmic ERß signaling may be more important for patient survival than its nuclear signaling. In the case of PR, positivity was an independent favorable prognostic factor for DFS. CONCLUSIONS: These novel findings, that need to be confirmed in a large prospective trial, suggest that additional prognostic, and possibly therapeutic opportunities may be available in advanced serous ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Estrogen Receptor beta/biosynthesis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Adult , Aged , Carboplatin/administration & dosage , Cell Nucleus/metabolism , Cisplatin/administration & dosage , Cohort Studies , Cystadenocarcinoma, Serous/pathology , Cytoplasm/metabolism , Disease-Free Survival , Estrogen Receptor alpha/biosynthesis , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Receptors, Progesterone/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...