Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Int J Radiat Biol ; 100(5): 744-755, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38466699

RESUMEN

PURPOSES: Lymphopenia is extensively studied, but not circulating leucocyte subpopulations, which however have distinct roles in tumor tolerance. Proton therapy has been shown to have a lesser impact on the immune system than conventional X-ray radiotherapy through lower dose exposure to healthy tissues. We explored the differential effects of brain X-ray and proton irradiation on circulating leucocyte subpopulations. MATERIALS AND METHODS: Leucocyte subpopulation counts from tumor-free mice were obtained 12 hours after 4 fractions of 2.5 Gy. The relationships between irradiation type (X-rays or protons), irradiated volume (whole-brain/hemi-brain) and dose rate (1 or 2 Gy/min) with circulating leucocyte subpopulations (T-CD4+, T-CD8+, B, and NK-cells, neutrophils, and monocytes) were investigated using linear regression and tree-based modeling approaches. Relationships between dose maps (brain, vessels, lymph nodes (LNs)) and leucocyte subpopulations were analyzed and applied to construct the blood dose model, assessing the hypothesis of a direct lymphocyte-killing effect in radiation-induced lymphopenia. RESULTS: Radiation-induced lymphopenia occurred after X-ray but not proton brain irradiation in lymphoid subpopulations (T-CD4+, T-CD8+, B, and NK-cells). There was an increase in neutrophil counts following protons but not X-rays. Monocytes remained unchanged under both X-rays and protons. Besides irradiation type, irradiated volume and dose rate had a significant impact on NK-cell, neutrophil and monocyte levels but not T-CD4+, T-CD8+, and B-cells. The dose to the blood had a heterogeneous impact on leucocyte subpopulations: neutrophil counts remained stable with increasing dose to the blood, while lymphocyte counts decreased with increasing dose (T-CD8+-cells > T-CD4+-cells > B-cells > NK-cells). Direct cell-killing effect of the dose to the blood mildly contributed to radiation-induced lymphopenia. LN exposure significantly contributed to lymphopenia and partially explained the distinct impact of irradiation type on circulating lymphocytes. CONCLUSIONS: Leucocyte subpopulations reacted differently to X-ray or proton brain irradiation. This difference could be partly explained by LN exposure to radiation dose. Further researches and analyses on other biological processes and interactions between leucocyte subpopulations are ongoing. The various mechanisms underlying leucocyte subpopulation changes under different irradiation modalities may have implications for the choice of radiotherapy modalities and their combination with immunotherapy in brain cancer treatment.


Asunto(s)
Encéfalo , Leucocitos , Animales , Ratones , Encéfalo/efectos de la radiación , Leucocitos/efectos de la radiación , Linfopenia/etiología , Relación Dosis-Respuesta en la Radiación , Masculino , Rayos X , Terapia de Protones/efectos adversos , Ratones Endogámicos C57BL
2.
Int J Radiat Oncol Biol Phys ; 118(4): 1081-1093, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-37866760

RESUMEN

PURPOSE: Radiation therapy for brain tumors increases patient survival. Nonetheless, side effects are increasingly reported such as cognitive deficits and fatigue. The etiology of fatigue remains poorly described. Our hypothesis is that the abscopal effects of radiation therapy on skeletal muscle may be involved in fatigue. The present study aims to assess the effect of brain irradiation on skeletal muscles and its relationship with fatigue and to analyze whether physical activity could counteract brain radiation-induced side effects. METHODS AND MATERIALS: Adult Wistar rats were randomly distributed between 4 groups: control (CTL), irradiated (IR), nonirradiated with physical activity (PA), and irradiated with physical activity (IR+PA). IR rats were exposed to a whole-brain irradiation (WBI) of 30 Gy (3 × 10 Gy). Rats subjected to PA underwent sessions of running on a treadmill, 3 times/week for 6 months. The effects of WBI on muscles were evaluated by complementary approaches: behavioral tests (fatigue, locomotion activity), magnetic resonance imaging, and histologic analyses. RESULTS: IR rats displayed a significant fatigue and a reduced locomotor activity at short term compared with the CTL group, which were attenuated with PA at 6 months after WBI. The IR rat's gastrocnemius mass decreased compared with CTL rats, which was reversed by physical activity at 14 days after WBI. Multiparametric magnetic resonance imaging of the skeletal muscle highlighted an alteration of the fiber organization in IR rats as demonstrated by a significant decrease of the mean diffusivity in the gastrocnemius at short term. Alteration of fibers was confirmed by histologic analyses: the number of type I fibers was decreased, whereas that of type IIa fibers was increased in IR animals but not in the IR+PA group. CONCLUSIONS: The data show that WBI induces skeletal muscle damage, which is attenuated by PA. This muscle damage may explain, at least in part, the fatigue of patients treated with radiation therapy.


Asunto(s)
Traumatismos por Radiación , Carrera , Humanos , Ratas , Animales , Ratas Wistar , Encéfalo/efectos de la radiación , Traumatismos por Radiación/etiología , Músculo Esquelético
3.
Am J Med Genet A ; 194(4): e63476, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37974505

RESUMEN

Cat Eye Syndrome (CES) is a rare genetic disease caused by the presence of a small supernumerary marker chromosome derived from chromosome 22, which results in a partial tetrasomy of 22p-22q11.21. CES is classically defined by association of iris coloboma, anal atresia, and preauricular tags or pits, with high clinical and genetic heterogeneity. We conducted an international retrospective study of patients carrying genomic gain in the 22q11.21 chromosomal region upstream from LCR22-A identified using FISH, MLPA, and/or array-CGH. We report a cohort of 43 CES cases. We highlight that the clinical triad represents no more than 50% of cases. However, only 16% of CES patients presented with the three signs of the triad and 9% not present any of these three signs. We also highlight the importance of other impairments: cardiac anomalies are one of the major signs of CES (51% of cases), and high frequency of intellectual disability (47%). Ocular motility defects (45%), abdominal malformations (44%), ophthalmologic malformations (35%), and genitourinary tract defects (32%) are other frequent clinical features. We observed that sSMC is the most frequent chromosomal anomaly (91%) and we highlight the high prevalence of mosaic cases (40%) and the unexpectedly high prevalence of parental transmission of sSMC (23%). Most often, the transmitting parent has mild or absent features and carries the mosaic marker at a very low rate (<10%). These data allow us to better delineate the clinical phenotype associated with CES, which must be taken into account in the cytogenetic testing for this syndrome. These findings draw attention to the need for genetic counseling and the risk of recurrence.


Asunto(s)
Aneuploidia , Trastornos de los Cromosomas , Cromosomas Humanos Par 22 , Anomalías del Ojo , Cardiopatías Congénitas , Humanos , Estudios Retrospectivos , Hibridación Fluorescente in Situ , Cromosomas Humanos Par 22/genética , Cardiopatías Congénitas/genética
4.
Cell Death Dis ; 14(12): 823, 2023 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-38092743

RESUMEN

The molecular mechanisms induced by hypoxia are misunderstood in non-small cell lung cancer (NSCLC), and above all the hypoxia and RASSF1A/Hippo signaling relationship. We confirmed that human NSCLC (n = 45) as their brain metastases (BM) counterpart are hypoxic since positive with CAIX-antibody (target gene of Hypoxia-inducible factor (HIF)). A severe and prolonged hypoxia (0.2% O2, 48 h) activated YAP (but not TAZ) in Human Bronchial Epithelial Cells (HBEC) lines by downregulating RASSF1A/kinases Hippo (except for NDR2) regardless their promoter methylation status. Subsequently, the NDR2-overactived HBEC cells exacerbated a HIF-1A, YAP and C-Jun-dependent-amoeboid migration, and mainly, support BM formation. Indeed, NDR2 is more expressed in human tumor of metastatic NSCLC than in human localized NSCLC while NDR2 silencing in HBEC lines (by shRNA) prevented the xenograft formation and growth in a lung cancer-derived BM model in mice. Collectively, our results indicated that NDR2 kinase is over-active in NSCLC by hypoxia and supports BM formation. NDR2 expression is thus a useful biomarker to predict the metastases risk in patients with NSCLC, easily measurable routinely by immunohistochemistry on tumor specimens.


Asunto(s)
Neoplasias Encefálicas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Humanos , Ratones , Neoplasias Encefálicas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Células Epiteliales/metabolismo , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/patología
5.
EJNMMI Res ; 13(1): 102, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-38006431

RESUMEN

BACKGROUND: Brain metastases (BM) are the most frequent malignant brain tumors. The aim of this study was to characterize the tumor microenvironment (TME) of BM and particularly hypoxia and redox state, known to play a role in tumor growth and treatment resistance with multimodal PET and MRI imaging, immunohistochemical and proteomic approaches in a human lung cancer (H2030-BrM3)-derived BM model in rats. RESULTS: First, in vitro studies confirmed that H2030-BrM3 cells respond to hypoxia with increasing expression of HIF-1, HIF-2 and their target genes. Proteomic analyses revealed, among expression changes, proteins associated with metabolism, oxidative stress, metal response and hypoxia signaling in particular in cortical BM. [64Cu][Cu(ATSM)] PET revealed a significant uptake by cortical BM (p < 0.01), while no uptake is observed in striatal BM 23 days after tumor implantation. Pimonidazole, HIF-1α, HIF-2α, CA-IX as well as GFAP, CTR1 and DMT1 immunostainings are positive in both BM. CONCLUSION: Overall, [64Cu][Cu(ATSM)] imaging and proteomic results showed the presence of hypoxia and protein expression changes linked to hypoxia and oxidative stress in BM, which are more pronounced in cortical BM compared to striatal BM. Moreover, it emphasized the interest of [64Cu][Cu(ATSM)] PET to characterize TME of BM and depict inter-metastasis heterogeneity that could be useful to guide treatments.

6.
NMR Biomed ; 36(3): e4858, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36285719

RESUMEN

Acute ischemic stroke results in an ischemic core surrounded by a tissue at risk, named the penumbra, which is potentially salvageable. One way to differentiate the tissues is to measure the hypoxia status. The purpose of the current study is to correlate the abnormal brain tissue volume derived from magnetic resonance-based imaging of brain oxygen saturation (St O2 -MRI) to the fluorine-18 fluoromisonidazole ([18 F]FMISO) positron emission tomography (PET) volume for hypoxia imaging validation, and to analyze the ability of St O2 -MRI to depict the different hypoxic tissue types in the acute phase of stroke. In a pertinent model of stroke in the rat, the volume of tissue with decreased St O2 -MRI signal and that with increased uptake of [18 F]FMISO were equivalent and correlated (r = 0.706; p = 0.015). The values of St O2 in the tissue at risk were significantly greater than those quantified in the core of the lesion, and were less than those for healthy tissue (52.3% ± 2.0%; 43.3% ± 1.9%, and 67.9 ± 1.4%, respectively). A threshold value for St O2 of ≈60% as the cut-off for the identification of the tissue at risk was calculated. Tissue volumes with reduced St O2 -MRI correlated with the final lesion (r = 0.964, p < 0.0001). The findings show that the St O2 -MRI approach is sensitive for the detection of hypoxia and for the prediction of the final lesion after stroke. Once validated in acute clinical settings, this approach might be used to enhance the stratification of patients for potential therapeutic interventions.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Ratas , Animales , Tomografía de Emisión de Positrones , Accidente Cerebrovascular/diagnóstico por imagen , Misonidazol , Hipoxia/diagnóstico por imagen , Imagen por Resonancia Magnética , Radiofármacos
7.
Clin Nucl Med ; 47(8): 699-706, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35485864

RESUMEN

OBJECTIVES: This study sought to assess 18 F-fludarabine ( 18 F-FLUDA) PET/CT's ability in differentiating primary central nervous system lymphomas (PCNSLs) from glioblastoma multiformes (GBMs). PATIENTS AND METHODS: Patients harboring either PCNSL (n = 8) before any treatment, PCNSL treated using corticosteroids (PCNSLh; n = 10), or GBM (n = 13) were investigated with conventional MRI and PET/CT, using 11 C-MET and 18 F-FLUDA. The main parameters measured with each tracer were SUV T and T/N ratios for the first 30 minutes of 11 C-MET acquisition, as well as at 3 different times after 18 F-FLUDA injection. The early 18 F-FLUDA uptake within the first minute of injection was equally considered, whereas this parameter was combined with the later uptakes to obtain R FLUDA 2 and R FLUDA 3 ratios. RESULTS: No significant differences in 11 C-MET uptakes were observed among PCNSL, PCNSLh, and GBM. With 18 F-FLUDA, a clear difference in dynamic GBM uptake was observed, which decreased over time after an early maximum, as compared with that of PCNSL, which steadily increased over time, PCNSLh exhibiting intermediate values. The most discriminative parameters consisting of R FLUDA 2 and R FLUDA 3 integrated the early tracer uptake (first 60 seconds), thereby provided 100% specificity and sensitivity. CONCLUSIONS: 18 F-FLUDA was shown to likely be a promising radiopharmaceutical for differentiating PCNSL from other malignancies, although a pretreatment with corticosteroids might compromise this differential diagnostic ability. The diagnostic role of 18 F-FLUDA should be further investigating, along with its potential of defining therapeutic strategies in patients with PCNSL, while assessing the treatments' effectiveness.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Linfoma , Corticoesteroides , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/patología , Diagnóstico Diferencial , Fluorodesoxiglucosa F18 , Glioblastoma/diagnóstico por imagen , Glioblastoma/patología , Humanos , Linfoma/diagnóstico por imagen , Linfoma/patología , Metionina , Tomografía Computarizada por Tomografía de Emisión de Positrones , Tomografía de Emisión de Positrones , Vidarabina/análogos & derivados
8.
Ann Biol Clin (Paris) ; 79(5): 427-438, 2021 Oct 01.
Artículo en Francés | MEDLINE | ID: mdl-34782309

RESUMEN

The aim of this study was to evaluate the ART outcomes for infertile males carrying a balanced structural chromosomal rearrangement or a Y-chromosome microdeletion, and to compare the results with a control group. The primary outcome was the clinical pregnancy rate. A retrospective case-control study has been carried out in the ART departments of the university hospitals of Bordeaux and la Réunion. Results of karyotypes and Y-chromosome microdeletions analysis of infertile men with sperm concentration lower than 5 millions/mL have been extracted from the softwares Jfiv® (Bordeaux) and MédiFirst® (la Réunion). The clinical pregnancy rate for carriers of balanced chromosomal rearrangements and Y-chromosome microdeletions was 28% and 43% respectively. The clinical pregnancy rate for the controls was 24% (non-significant difference). According to this study, balanced chromosomal rearrangements and Y-chromosome microdeletions are not found to affect the clinical pregnancy rate in conventional ART. Further larger scale studies are required to confirm these results.


Asunto(s)
Azoospermia , Infertilidad Masculina , Azoospermia/genética , Estudios de Casos y Controles , Deleción Cromosómica , Cromosomas Humanos Y/genética , Femenino , Humanos , Infertilidad Masculina/genética , Masculino , Embarazo , Técnicas Reproductivas Asistidas , Estudios Retrospectivos , Aberraciones Cromosómicas Sexuales , Trastornos de los Cromosomas Sexuales del Desarrollo Sexual
9.
Nucleic Acids Res ; 49(19): 11241-11256, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34634812

RESUMEN

The stable insertion of the retroviral genome into the host chromosomes requires the association between integration complexes and cellular chromatin via the interaction between retroviral integrase and the nucleosomal target DNA. This final association may involve the chromatin-binding properties of both the retroviral integrase and its cellular cofactor LEDGF/p75. To investigate this and better understand the LEDGF/p75-mediated chromatin tethering of HIV-1 integrase, we used a combination of biochemical and chromosome-binding assays. Our study revealed that retroviral integrase has an intrinsic ability to bind and recognize specific chromatin regions in metaphase even in the absence of its cofactor. Furthermore, this integrase chromatin-binding property was modulated by the interaction with its cofactor LEDGF/p75, which redirected the enzyme to alternative chromosome regions. We also better determined the chromatin features recognized by each partner alone or within the functional intasome, as well as the chronology of efficient LEDGF/p75-mediated targeting of HIV-1 integrase to chromatin. Our data support a new chromatin-binding function of integrase acting in concert with LEDGF/p75 for the optimal association with the nucleosomal substrate. This work also provides additional information about the behavior of retroviral integration complexes in metaphase chromatin and the mechanism of action of LEDGF/p75 in this specific context.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cromatina/metabolismo , Integrasa de VIH/genética , Histonas/genética , Interacciones Huésped-Patógeno/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Cromatina/química , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Integrasa de VIH/metabolismo , Histonas/metabolismo , Humanos , Células K562 , Cultivo Primario de Células , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T/virología , Factores de Transcripción/metabolismo
10.
Sci Rep ; 11(1): 11239, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045576

RESUMEN

Lung cancer patients frequently develop brain metastases (BM). Despite aggressive treatment including neurosurgery and external-radiotherapy, overall survival remains poor. There is a pressing need to further characterize factors in the microenvironment of BM that may confer resistance to radiotherapy (RT), such as hypoxia. Here, hypoxia was first evaluated in 28 biopsies from patients with non­small cell lung cancer (NSCLC) BM, using CA-IX immunostaining. Hypoxia characterization (pimonidazole, CA-IX and HIF-1α) was also performed in different preclinical NSCLC BM models induced either by intracerebral injection of tumor cells (H2030-Br3M, H1915) into the cortex and striatum, or intracardial injection of tumor cells (H2030-Br3M). Additionally, [18F]-FMISO-PET and oxygen-saturation-mapping-MRI (SatO2-MRI) were carried out in the intracerebral BM models to further characterize tumor hypoxia and evaluate the potential of Hypoxia-image-guided-RT (HIGRT). The effect of RT on proliferation of BM ([18F]-FLT-PET), tumor volume and overall survival was determined. We showed that hypoxia is a major yet heterogeneous feature of BM from lung cancer both preclinically and clinically. HIGRT, based on hypoxia heterogeneity observed between cortical and striatal metastases in the intracerebrally induced models, showed significant potential for tumor control and animal survival. These results collectively highlight hypoxia as a hallmark of BM from lung cancer and the value of HIGRT in better controlling tumor growth.


Asunto(s)
Neoplasias Encefálicas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Neoplasias Pulmonares/diagnóstico por imagen , Radioterapia Guiada por Imagen , Hipoxia Tumoral , Anciano , Animales , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Carcinoma de Pulmón de Células no Pequeñas/secundario , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Imagen por Resonancia Magnética , Persona de Mediana Edad , Ratas , Sistema de Registros
11.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33558238

RESUMEN

Propranolol, a nonselective ß-adrenergic receptor (ADRB) antagonist, is the first-line therapy for severe infantile hemangiomas (IH). Since the incidental discovery of propranolol efficacy in IH, preclinical and clinical investigations have shown evidence of adjuvant propranolol response in some malignant tumors. However, the mechanism for propranolol antitumor effect is still largely unknown, owing to the absence of a tumor model responsive to propranolol at nontoxic concentrations. Immunodeficient mice engrafted with different human tumor cell lines were treated with anti-VEGF bevacizumab to create a model sensitive to propranolol. Proteomics analysis was used to reveal propranolol-mediated protein alteration correlating with tumor growth inhibition, and Aquaporin-1 (AQP1), a water channel modulated in tumor cell migration and invasion, was identified. IH tissues and cells were then functionally investigated. Our functional protein association networks analysis and knockdown of ADRB2 and AQP1 indicated that propranolol treatment and AQP1 down-regulation trigger the same pathway, suggesting that AQP1 is a major driver of beta-blocker antitumor response. Examining AQP1 in human hemangioma samples, we found it exclusively in a perivascular layer, so far unrecognized in IH, made of telocytes (TCs). Functional in vitro studies showed that AQP1-positive TCs play a critical role in IH response to propranolol and that modulation of AQP1 in IH-TC by propranolol or shAQP1 decreases capillary-like tube formation in a Matrigel-based angiogenesis assay. We conclude that IH sensitivity to propranolol may rely, at least in part, on a cross talk between lesional vascular cells and stromal TCs.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Acuaporina 1/metabolismo , Hemangioma Capilar/metabolismo , Síndromes Neoplásicos Hereditarios/metabolismo , Neovascularización Patológica/metabolismo , Propranolol/farmacología , Telocitos/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Hemangioma Capilar/tratamiento farmacológico , Humanos , Ratones , Síndromes Neoplásicos Hereditarios/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Propranolol/uso terapéutico , Proteoma/genética , Proteoma/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Telocitos/efectos de los fármacos , Telocitos/fisiología
12.
Stem Cell Reports ; 15(3): 677-693, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32795423

RESUMEN

CRISPR/Cas9 is a promising technology for gene correction. However, the edition is often biallelic, and uncontrolled small insertions and deletions (indels) concomitant to precise correction are created. Mutation-specific guide RNAs were recently tested to correct dominant inherited diseases, sparing the wild-type allele. We tested an original approach to correct compound heterozygous recessive mutations. We compared editing efficiency and genotoxicity by biallelic guide RNA versus mutant allele-specific guide RNA in iPSCs derived from a congenital erythropoietic porphyria patient carrying compound heterozygous mutations resulting in UROS gene invalidation. We obtained UROS function rescue and metabolic correction with both guides with the potential of use for porphyria clinical intervention. However, unlike the biallelic one, the mutant allele-specific guide was free of on-target collateral damage. We recommend this design to avoid genotoxicity and to obtain on-target scarless gene correction for recessive disease with frequent cases of compound heterozygous mutations.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Sistemas CRISPR-Cas/genética , Edición Génica , Mutación/genética , Porfirias/genética , Porfirias/terapia , ARN Guía de Kinetoplastida/metabolismo , Células Madre/metabolismo , Alelos , Secuencia de Bases , Células Clonales , Exones/genética , Terapia Genética , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Cariotipificación , Uroporfirinógeno III Sintetasa/genética
13.
Biomaterials ; 257: 120249, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32739663

RESUMEN

Approaches able to counteract, at least temporarily, hypoxia, a well-known factor of resistance to treatment in solid tumors are highly desirable. Herein, we report the use of nanosized zeolite crystals as hyperoxic/hypercapnic gas carriers for glioblastoma. First, the non-toxic profile of nanosized zeolite crystals in living animals (mice, rats and non-human primates) and in various cell types is presented. Second, the ability of the nanosized zeolites to act as a vasoactive agent for a targeted re-oxygenation of the tumor after intravenous injection is shown. As attested by an MRI protocol, the zeolites were able to increase oxygenation and blood volume specifically within the brain tumor whilst no changes in the healthy-non tumoral brain-were observed. The first proof of concept for the use of metal-containing nanosized zeolites as a tool for vectorization of hyperoxic/hypercapnic gases in glioblastoma is revealed.


Asunto(s)
Glioblastoma , Zeolitas , Animales , Gases , Imagen por Resonancia Magnética , Ratones , Ratas
14.
Cancers (Basel) ; 12(8)2020 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-32718037

RESUMEN

Tumor hypoxia is known to limit the efficacy of ionizing radiations, a concept called oxygen enhancement ratio (OER). OER depends on physical factors such as pO2 and linear energy transfer (LET). Biological pathways, such as the hypoxia-inducible transcription factors (HIF), might also modulate the influence of LET on OER. Glioblastoma (GB) is resistant to low-LET radiation (X-rays), due in part to the hypoxic environment in this brain tumor. Here, we aim to evaluate in vitro whether high-LET particles, especially carbon ion radiotherapy (CIRT), can overcome the contribution of hypoxia to radioresistance, and whether HIF-dependent genes, such as erythropoietin (EPO), influence GB sensitivity to CIRT. Hypoxia-induced radioresistance was studied in two human GB cells (U251, GL15) exposed to X-rays or to carbon ion beams with various LET (28, 50, 100 keV/µm), and in genetically-modified GB cells with downregulated EPO signaling. Cell survival, radiobiological parameters, cell cycle, and ERK activation were assessed under those conditions. The results demonstrate that, although CIRT is more efficient than X-rays in GB cells, hypoxia can limit CIRT efficacy in a cell-type manner that may involve differences in ERK activation. Using high-LET carbon beams, or targeting hypoxia-dependent genes such as EPO might reduce the effects of hypoxia.

16.
Neuro Oncol ; 22(3): 357-368, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31538194

RESUMEN

BACKGROUND: Brain metastases (BM) develop frequently in patients with breast cancer. Despite the use of external beam radiotherapy (EBRT), the average overall survival is short (6 months from diagnosis). The therapeutic challenge is to deliver molecularly targeted therapy at an early stage when relatively few metastatic tumor cells have invaded the brain. Vascular cell adhesion molecule 1 (VCAM-1), overexpressed by nearby endothelial cells during the early stages of BM development, is a promising target. The aim of this study was to investigate the therapeutic value of targeted alpha-particle radiotherapy, combining lead-212 (212Pb) with an anti-VCAM-1 antibody (212Pb-αVCAM-1). METHODS: Human breast carcinoma cells that metastasize to the brain, MDA-231-Br-GFP, were injected into the left cardiac ventricle of nude mice. Twenty-one days after injection, 212Pb-αVCAM-1 uptake in early BM was determined in a biodistribution study and systemic/brain toxicity was evaluated. Therapeutic efficacy was assessed using MR imaging and histology. Overall survival after 212Pb-αVCAM-1 treatment was compared with that observed after standard EBRT. RESULTS: 212Pb-αVCAM-1 was taken up into early BM with a tumor/healthy brain dose deposition ratio of 6 (5.52e108 and 0.92e108) disintegrations per gram of BM and healthy tissue, respectively. MRI analyses showed a statistically significant reduction in metastatic burden after 212Pb-αVCAM-1 treatment compared with EBRT (P < 0.001), translating to an increase in overall survival of 29% at 40 days post prescription (P < 0.01). No major toxicity was observed. CONCLUSIONS: The present investigation demonstrates that 212Pb-αVCAM-1 specifically accumulates at sites of early BM causing tumor growth inhibition.


Asunto(s)
Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Radioterapia/métodos , Molécula 1 de Adhesión Celular Vascular/inmunología , Partículas alfa , Animales , Anticuerpos/administración & dosificación , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Femenino , Humanos , Radioisótopos de Plomo/administración & dosificación , Ratones , Ratones Desnudos
17.
Med Phys ; 47(1): 234-241, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31688950

RESUMEN

PURPOSE: New preclinical image-guided irradiators and treatment planning systems represent a huge progress in radiobiology. Nevertheless, quality control of preclinical treatments is not as advanced as in clinical radiotherapy and in vivo dosimetry is less developed. In this study, we evaluate the use of a scintillating fiber dosimeter called DosiRat to verify the agreement between the doses planned with SmART-Plan and the measured doses during small animal irradiations. METHODS: In vivo dosimetry was first evaluated with DosiRat through dose measurements performed at the surface of a 3 × 9 × 3 cm3 phantom. Measured and planned doses were compared for different irradiation conditions (prescription point, anterior, and posterior beams, 5 mm and 10 mm irradiation fields). In a second phase, measured and planned doses were compared for rat brain irradiations performed with anterior beams, with DosiRat positioned at the beam entrance. Comparisons were performed for different tube currents (1.3 and 13 mA), collimations (5, 10 and 25 mm diameter), and planned doses (0.1, 0.5, 2, and 10 Gy). RESULTS: In the case of the phantom irradiations, planned and measured doses showed discrepancies smaller than the 5% accuracy of the TPS, except in cases in which the dosimeter was not centered in the irradiation field. The differences were larger for animal irradiations (from -3.3% to 8.8%) because of variations of the beam energy spectrum and the nonequivalence between materials at medium and low energy. CONCLUSIONS: This study highlighted the complexity to implement one-dimension in vivo dosimetry in orthovoltage millimetric beams. Nevertheless, DosiRat is well adapted to in vivo dosimetry because of its small volume and its direct reading and allowed in vivo control of planned doses for anterior beams down to 5 mm diameter.


Asunto(s)
Radiometría/instrumentación , Radioterapia Guiada por Imagen/instrumentación , Conteo por Cintilación/instrumentación , Animales , Encéfalo/efectos de la radiación , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador , Ratas
18.
EJNMMI Res ; 9(1): 114, 2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31858290

RESUMEN

BACKGROUND: Diacetyl-bis(N4-methylthiosemicarbazone), labeled with 64Cu (64Cu-ATSM) has been suggested as a promising tracer for imaging hypoxia. However, various controversial studies highlighted potential pitfalls that may disable its use as a selective hypoxic marker. They also highlighted that the results may be tumor location dependent. Here, we first analyzed uptake of Cu-ATSM and its less lipophilic counterpart Cu-Cl2 in the tumor over time in an orthotopic glioblastoma model. An in vitro study was also conducted to investigate the hypoxia-dependent copper uptake in tumor cells. We then further performed a comprehensive ex vivo study to compare 64Cu uptake to hypoxic markers, specific cellular reactions, and also transporter expression. METHODS: µPET was performed 14 days (18F-FMISO), 15 days (64Cu-ATSM and 64Cu-Cl2), and 16 days (64Cu-ATSM and 64Cu-Cl2) after C6 cell inoculation. Thereafter, the brains were withdrawn for further autoradiography and immunohistochemistry. C6 cells were also grown in hypoxic workstation to analyze cellular uptake of Cu complexes in different oxygen levels. RESULTS: In vivo results showed that Cu-ASTM and Cu-Cl2 accumulated in hypoxic areas of the tumors. Cu-ATSM also stained, to a lesser extent, non-hypoxic regions, such as regions of astrogliosis, with high expression of copper transporters and in particular DMT-1 and CTR1, and also characterized by the expression of elevated astrogliosis. In vitro results show that 64Cu-ATSM showed an increase in the uptake only in severe hypoxia at 0.5 and 0.2% of oxygen while for 64Cu-Cl2, the cell retention was significantly increased at 5% and 1% of oxygen with no significant rise at lower oxygen percentages. CONCLUSION: In the present study, we show that Cu-complexes undoubtedly accumulate in hypoxic areas of the tumors. This uptake may be the reflection of a direct dependency to a redox metabolism and also a reflection of hypoxic-induced overexpression of transporters. We also show that Cu-ATSM also stained non-hypoxic regions such as astrogliosis.

19.
Nat Commun ; 10(1): 1136, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30850590

RESUMEN

CRISPR-Cas9 is a promising technology for genome editing. Here we use Cas9 nuclease-induced double-strand break DNA (DSB) at the UROS locus to model and correct congenital erythropoietic porphyria. We demonstrate that homology-directed repair is rare compared with NHEJ pathway leading to on-target indels and causing unwanted dysfunctional protein. Moreover, we describe unexpected chromosomal truncations resulting from only one Cas9 nuclease-induced DSB in cell lines and primary cells by a p53-dependent mechanism. Altogether, these side effects may limit the promising perspectives of the CRISPR-Cas9 nuclease system for disease modeling and gene therapy. We show that the single nickase approach could be safer since it prevents on- and off-target indels and chromosomal truncations. These results demonstrate that the single nickase and not the nuclease approach is preferable, not only for modeling disease but also and more importantly for the safe management of future CRISPR-Cas9-mediated gene therapies.


Asunto(s)
Sistemas CRISPR-Cas , Cromosomas Humanos Par 10 , Roturas del ADN de Doble Cadena , Desoxirribonucleasa I/genética , Edición Génica/métodos , Terapia Genética/métodos , Uroporfirinógeno III Sintetasa/genética , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Deleción Cromosómica , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , ADN/genética , ADN/metabolismo , Desoxirribonucleasa I/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Genoma Humano , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Células K562 , Modelos Biológicos , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/metabolismo , Porfiria Eritropoyética/patología , Porfiria Eritropoyética/terapia , Cultivo Primario de Células , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Reparación del ADN por Recombinación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Uroporfirinógeno III Sintetasa/metabolismo
20.
Clin Genet ; 95(3): 420-426, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30633342

RESUMEN

Rubinstein-Taybi syndrome (RSTS; OMIM 180849) is an autosomal dominant developmental disorder characterized by facial dysmorphism, broad thumbs and halluces associated with intellectual disability. RSTS is caused by alterations in CREBBP (about 60%) and EP300 genes (8%). RSTS is often diagnosed at birth or during early childhood but generally not suspected during antenatal period. We report nine cases of well-documented fetal RSTS. Two cases were examined after death in utero at 18 and 35 weeks of gestation and seven cases after identification of ultrasound abnormalities and termination of pregnancy. On prenatal sonography, a large gallbladder was detected in two cases, and brain malformations were noted in four cases, especially cerebellar hypoplasia. However, the diagnosis of RSTS has not been suggested during pregnancy. Fetal autopsy showed that all fetuses had large thumbs and/or suggestive facial dysmorphism. A CREBBP gene anomaly was identified in all cases. Alterations were similar to those found in typical RSTS children. This report will contribute to a better knowledge of the fetal phenotype to consider the hypothesis of RSTS during pregnancy. Genotyping allows reassuring genetic counseling.


Asunto(s)
Proteína de Unión a CREB/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Fenotipo , Síndrome de Rubinstein-Taybi/diagnóstico , Síndrome de Rubinstein-Taybi/genética , Autopsia , Femenino , Muerte Fetal , Dosificación de Gen , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Masculino , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA