Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Probl Radiac Med Radiobiol ; 24: 65-92, 2019 Dec.
Article in English, Ukrainian | MEDLINE | ID: mdl-31841459

ABSTRACT

The review summarizes and analyzes the data of world scientific literature and the results of the own research con- cerning one of the main non-targeted effects of ionizing radiation - the radiation induced bystander effect (RIBE) - the ability of irradiated target cells to induce secondary biological changes in non-irradiated receptor cells. The his- tory of studies of this phenomenon is presented - it described under various names since 1905, began to study from the end of the twentieth century when named as RIBE and caused particular interest in the scientific community during recent decades. It is shown that the development of biological science and the improvement of research methods allowed to get new in-depth data on the development of RIBE not only at the level of the whole organism, but even at the genome level. The review highlights the key points of numerous RIBE investigations including mod- eling; methodological approaches to studying; classification; features of interaction between irradiated and intact cells; the role of the immune system, oxidative stress, cytogenetic disorders, changes in gene expression in the mechanism of development of RIBE; rescue effect, abscopal effect, persistence, modification, medical effects. It is emphasized that despite the considerable amount of research concerning the bystander response as the universal phenomenon and RIBE as one of its manifestations, there are still enough «white spots¼ in determining the mech- anisms of the RIBE formation and assessing the possible consequences of its development for human health.


Subject(s)
Bystander Effect/radiation effects , Leukemia, Radiation-Induced/pathology , Models, Biological , Neoplasms, Radiation-Induced/pathology , Radiation, Ionizing , Animals , Apoptosis/immunology , Apoptosis/radiation effects , Bystander Effect/genetics , Bystander Effect/immunology , Cytokines/biosynthesis , Genomic Instability/immunology , Genomic Instability/radiation effects , Humans , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/immunology , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/immunology
2.
Health Phys ; 109(5): 414-26, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26425902

ABSTRACT

Recovery from severe immunosuppression requires hematopoietic stem cell reconstitution and effective thymopoiesis to restore a functional immune cell repertoire. Herein, a model of immune cell reconstitution consequent to potentially lethal doses of irradiation is described, which may be valuable in evaluating potential medical countermeasures. Male rhesus macaques were total body irradiated by exposure to 6.00 Gy 250 kVp x-radiation (midline tissue dose, 0.13 Gy min), resulting in an approximate LD10/60 (n = 5/59). Animals received medical management, and hematopoietic and immune cell recovery was assessed (n ≤ 14) through 370 d post exposure. A subset of animals (n ≤ 8) was examined through 700 d. Myeloid recovery was assessed by neutrophil and platelet-related parameters. Lymphoid recovery was assessed by the absolute lymphocyte count and FACS-based phenotyping of B- and T-cell subsets. Recent thymic emigrants were identified by T cell receptor excision circle quantification. Severe neutropenia, lymphopenia, and thrombocytopenia resolved within 30 d. Total CD3+ cells µL required 60 d to reach values 60% of normal, followed by subsequent slow recovery to approximately normal by 180 d post irradiation. Recovery of CD3+4+ and CD3+8+ cell memory and naïve subsets were markedly different. Memory populations were ≥ 100% of normal by day 60, whereas naïve populations were only 57% normal at 180 d and never fully recovered to baseline post irradiation. Total (CD20+) B cells µL were within normal levels by 77 d post exposure. This animal model elucidates the variable T- and B-cell subset recovery kinetics after a potentially lethal dose of total-body irradiation that are dependent on marrow-derived stem and progenitor cell recovery, peripheral homeostatic expansion, and thymopoiesis.


Subject(s)
Immune Reconstitution Inflammatory Syndrome/immunology , Leukemia, Radiation-Induced/etiology , Leukemia, Radiation-Induced/immunology , Lymphocytes/immunology , Myeloid Cells/immunology , Recovery of Function/immunology , Animals , Immune Reconstitution Inflammatory Syndrome/pathology , Leukemia, Radiation-Induced/pathology , Lymphocytes/radiation effects , Macaca mulatta , Male , Myeloid Cells/radiation effects , Radiation Dosage , Recovery of Function/physiology , Whole-Body Irradiation/adverse effects , X-Rays
3.
PLoS One ; 9(7): e104819, 2014.
Article in English | MEDLINE | ID: mdl-25126721

ABSTRACT

Estimates of cancer risks posed to space-flight crews by exposure to high atomic number, high-energy (HZE) ions are subject to considerable uncertainty because epidemiological data do not exist for human populations exposed to similar radiation qualities. We assessed the carcinogenic effects of 300 MeV/n 28Si or 600 MeV/n 56Fe ions in a mouse model for radiation-induced acute myeloid leukemia and hepatocellular carcinoma. C3H/HeNCrl mice were irradiated with 0.1, 0.2, 0.4, or 1 Gy of 300 MeV/n 28Si ions, 600 MeV/n 56Fe ions or 1 or 2 Gy of protons simulating the 1972 solar particle event (1972SPE) at the NASA Space Radiation Laboratory. Additional mice were irradiated with 137Cs gamma rays at doses of 1, 2, or 3 Gy. All groups were followed until they were moribund or reached 800 days of age. We found that 28Si or 56Fe ions do not appear to be substantially more effective than gamma rays for the induction of acute myeloid leukemia. However, 28Si or 56Fe ion irradiated mice had a much higher incidence of hepatocellular carcinoma than gamma ray irradiated or proton irradiated mice. These data demonstrate a clear difference in the effects of these HZE ions on the induction of leukemia compared to solid tumors, suggesting potentially different mechanisms of tumorigenesis. Also seen in this study was an increase in metastatic hepatocellular carcinoma in the 28Si and 56Fe ion irradiated mice compared with those exposed to gamma rays or 1972SPE protons, a finding with important implications for setting radiation exposure limits for space-flight crew members.


Subject(s)
Carcinoma, Hepatocellular/etiology , Cosmic Radiation/adverse effects , Leukemia, Myeloid, Acute/etiology , Leukemia, Radiation-Induced/etiology , Liver Neoplasms, Experimental/etiology , Radiation Injuries, Experimental/etiology , Animals , Carcinoma, Hepatocellular/secondary , Humans , Iron/adverse effects , Leukemia, Myeloid, Acute/pathology , Leukemia, Radiation-Induced/pathology , Liver Neoplasms, Experimental/pathology , Male , Mice, Inbred C3H , Radiation Injuries, Experimental/pathology , Silicon/adverse effects , Space Flight
4.
Int J Radiat Oncol Biol Phys ; 90(1): 53-62, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24974217

ABSTRACT

PURPOSE: Ionizing radiation is a common carcinogen that is important for the development of leukemia. However, the underlying epigenetic mechanisms remain largely unknown. The goal of the study was to explore microRNAome alterations induced by ionizing radiation (IR) in murine thymus, and to determine the role of IR-inducible microRNA (miRNA/miR) in the development of leukemia. METHODS AND MATERIALS: We used the well-established C57BL/6 mouse model and miRNA microarray profiling to identify miRNAs that are differentially expressed in murine thymus in response to irradiation. TIB152 human leukemia cell line was used to determine the role of estrogen receptor-α (ERα) in miR-27b transcription. The biological effects of ectopic miR-27b on leukemogenesis were measured by western immunoblotting, cell viability, apoptosis, and cell cycle analyses. RESULTS: Here, we have shown that IR triggers the differential expression of miR-27b in murine thymus tissue in a dose-, time- and sex-dependent manner. miR-27b was significantly down-regulated in leukemia cell lines CCL119 and TIB152. Interestingly, ERα was overexpressed in those 2 cell lines, and it was inversely correlated with miR-27b expression. Therefore, we used TIB152 as a model system to determine the role of ERα in miR-27b expression and the contribution of miR-27b to leukemogenesis. ß-Estradiol caused a rapid and transient reduction in miR-27b expression reversed by either ERα-neutralizing antibody or ERK1/2 inhibitor. Ectopic expression of miR-27b remarkably suppressed TIB152 cell proliferation, at least in part, by inducing S-phase arrest. In addition, it attenuated the expression of cyclin A2, although it had no effect on the levels of PCNA, PPARγ, CDK2, p21, p27, p-p53, and cleaved caspase-3. CONCLUSION: Our data reveal that ß-estradiol/ERα signaling may contribute to the down-regulation of miR-27b in acute leukemia cell lines through the ERK1/2 pathway, and that miR-27b may function as a tumor suppressor that inhibits cell proliferation by targeting cyclin A2.


Subject(s)
Leukemia, Radiation-Induced/metabolism , MicroRNAs/metabolism , Receptors, Estrogen/metabolism , Thymus Gland/radiation effects , Animals , Cell Line, Tumor , Cell Proliferation , Cyclin A2/metabolism , Dose-Response Relationship, Radiation , Down-Regulation , Estradiol/metabolism , Estradiol/pharmacology , Female , Humans , Leukemia, Radiation-Induced/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Estrogen/antagonists & inhibitors , S Phase Cell Cycle Checkpoints , Sex Factors , Thymus Gland/drug effects , Thymus Gland/metabolism , Time Factors
5.
Clin Lymphoma Myeloma Leuk ; 14(5): 401-10, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24875590

ABSTRACT

INTRODUCTION/BACKGROUND: We evaluated the characteristics of a cohort of patients with myelodysplastic syndrome (MDS) related to therapy (t-MDS) to create a prognostic model. PATIENTS AND METHODS: We identified 281 patients with MDS who had received previous chemotherapy and/or radiotherapy for previous malignancy. Potential prognostic factors were determined using univariate and multivariate analyses. RESULTS: Multivariate Cox regression analysis identified 7 factors that independently predicted short survival in t-MDS: age ≥ 65 years (hazard ratio [HR], 1.63), Eastern Cooperative Oncology Group performance status 2-4 (HR, 1.86), poor cytogenetics (-7 and/or complex; HR, 2.47), World Health Organization MDS subtype (RARs or RAEB-1/2; HR, 1.92), hemoglobin (< 11 g/dL; HR, 2.24), platelets (< 50 × 10(9)/dL; HR, 2.01), and transfusion dependency (HR, 1.59). These risk factors were used to create a prognostic model that segregated patients into 3 groups with distinct median overall survival: good (0-2 risk factors; 34 months), intermediate (3-4 risk factors; 12 months), and poor (5-7 risk factors; 5 months) (P < .001) and 1-year leukemia-free survival (96%, 84%, and 72%, respectively, P = .003). This model also identified distinct survival groups according to t-MDS therapy. CONCLUSION: In summary, we devised a prognostic model specifically for patients with t-MDS that predicted overall survival and leukemia-free survival. This model might facilitate the development of risk-adapted therapeutic strategies.


Subject(s)
Antineoplastic Agents/adverse effects , Leukemia, Myeloid, Acute/etiology , Leukemia, Radiation-Induced/etiology , Models, Biological , Myelodysplastic Syndromes/etiology , Neoplasms, Second Primary/etiology , Radiotherapy/adverse effects , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Blood Transfusion/statistics & numerical data , Disease Progression , Disease-Free Survival , Female , Hematopoietic Stem Cell Transplantation/adverse effects , Hemoglobins/analysis , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Radiation-Induced/mortality , Leukemia, Radiation-Induced/pathology , Male , Middle Aged , Multivariate Analysis , Myelodysplastic Syndromes/chemically induced , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/pathology , Neoplasms, Second Primary/chemically induced , Neoplasms, Second Primary/mortality , Platelet Count , Prognosis , Proportional Hazards Models , Risk Factors , Young Adult
6.
Radiat Res ; 178(5): 489-98, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23050984

ABSTRACT

Leukemia is one of the earliest cancer effects observed after acute exposure to relatively high doses of ionizing radiation. Leukemia mortality after external exposure at low doses and low-dose rates has been investigated at the French Atomic Energy Commission (CEA) and Nuclear Fuel Company (AREVA NC) after an additional follow-up of 10 years. The cohort included radiation-monitored workers employed for at least one year during 1950-1994 at CEA or AREVA NC and followed during 1968-2004. Association between external exposure and leukemia mortality was estimated with excess relative risk (ERR) models and time-dependent modifying factors were investigated with time windows. The cohort included 36,769 workers, followed for an average of 28 years, among whom 73 leukemia deaths occurred. Among the workers with a positive recorded dose, the mean cumulative external dose was 21.7 mSv. Results under a 2-year lag assumption suggested that the risk of leukemia (except chronic lymphatic leukemia) increased significantly by 8% per 10 mSv. The magnitude of the association for myeloid leukemia was larger. The higher ERR/Sv for doses received 2-14 years earlier suggest that time since exposure modifies the effect. The ERR/Sv also appeared higher for doses received at exposure rates ≥20 mSv per year. These results are consistent with those found in other studies of nuclear workers. However, confidence intervals are still wide. Further analyses should be conducted in pooled cohorts of nuclear workers.


Subject(s)
Leukemia, Radiation-Induced/mortality , Occupational Exposure , Radiation, Ionizing , Dose-Response Relationship, Radiation , Female , Follow-Up Studies , France , Humans , Leukemia, Radiation-Induced/pathology , Male , Middle Aged , Nuclear Energy , Nuclear Reactors , Risk Factors
7.
Article in English | MEDLINE | ID: mdl-22852813

ABSTRACT

Despite the fact that animal and human epidemiological studies confirmed a link between radon exposure in homes and increased risk of lung cancer in general population, other types of cancers induced by radon, such as leukemia, have not been consistently demonstrated. The aim of this review was to summarize data published thus far from ecological and case-control studies in exposed populations, taking into account radon dose estimation and evidence of radon-induced genotoxicity, in an effort to clarify the correlation between home radon exposure and incidence of childhood leukemia. Among 12 ecological studies, 11 reported a positive association between radon levels and elevated frequency of childhood leukemia, with 8 being significant. In conjunction with ecological studies, several case-control studies on indoor radon exposure and childhood leukemia were examined, and most investigations indicated a weak association with only a few showing significance. A major source of uncertainty in radon risk assessment is radon dose estimate. Methods for radon exposure measurement in homes of children are one of the factors that affect the risk estimates in a case-control study. The effects of radon-induced genetic damage were studied both in vitro and in vivo using genetic endpoints including chromosomal aberration (CA), micronuclei (MN) formation, gene mutation, and deletions and insertions. By applying a meta-analysis, an increased risk of childhood leukemia induced by indoor radon exposure was noted for overall leukemia and for acute lymphoblastic leukemia (ALL). Data thus indicated an association between environmental radon exposure and elevated leukemia incidence, but more evidence is required in both human investigations and animal mechanistic research before this assumption may be confirmed with certainty.


Subject(s)
Air Pollution, Indoor/adverse effects , Environmental Exposure/adverse effects , Leukemia, Radiation-Induced/epidemiology , Radon/toxicity , Animals , Child , Environmental Monitoring/methods , Epidemiological Monitoring , Humans , Incidence , Leukemia, Radiation-Induced/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/etiology , Radiation Dosage , Radon/analysis , Risk Assessment , Risk Factors
8.
Carcinogenesis ; 33(11): 2269-75, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22843505

ABSTRACT

H-ferritin (HF) is a core subunit of the iron storage protein ferritin and is related to the pathogenesis of malignant diseases. HF overexpression is present in human hematologic malignancies, suggesting that HF overexpression may contribute to the development of hematologic cancers. However, in vivo evidence that HF is directly linked to hematologic tumorigenesis has not yet been shown. In this study, we show that transgenic (tg) mice overexpressing the human HF gene (hHF-tg) developed aggressive radiation-induced thymic lymphoma/leukemia (TL) compared with wild-type (WT) mice, providing evidence that HF overexpression promotes leukemia/lymphomagenesis. Fractionated X-irradiation of hHF-tg mice caused a higher incidence and earlier onset of TL compared with WT mice. Immunological and pathological features of TLs were similar in both groups. However, proliferative activity of hHF-tg lymphoma cells was higher than that of WT lymphoma cells, and microarray analyses revealed that some leukemia/lymphoma-related genes were differentially expressed in hHF-tg TLs compared with WT TLs. To investigate whether cell damage induced by irradiation is related to leukemia/lymphomagenesis, we evaluated apoptotic levels in the thymus and bone marrow (BM) of hHF-tg and WT groups after fractionated X-irradiation. Apoptosis was augmented in the hHF-tg BM, but not in the thymus, compared with the WT BM, suggesting a possible linkage between increased BM apoptosis by HF overexpression and accelerated radiation-induced TL development. Our findings indicate that HF overexpression is closely related to the development of leukemia/lymphoma, which could have implications for the prevention of malignant hematologic diseases.


Subject(s)
Apoferritins/physiology , Apoptosis/radiation effects , Leukemia, Radiation-Induced/etiology , Lymphoma/etiology , X-Rays/adverse effects , 8-Hydroxy-2'-Deoxyguanosine , Animals , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Flow Cytometry , Humans , Leukemia, Radiation-Induced/mortality , Leukemia, Radiation-Induced/pathology , Lymphoma/mortality , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Survival Rate
9.
Br J Haematol ; 155(2): 167-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21810091

ABSTRACT

NUP98-HOXA9 [t(7;11) (p15;p15)] is associated with inferior prognosis in de novo and treatment-related acute myeloid leukaemia (AML) and contributes to blast crisis in chronic myeloid leukaemia (CML). We have engineered an inducible transgenic zebrafish harbouring human NUP98-HOXA9 under the zebrafish spi1(pu.1) promoter. NUP98-HOXA9 perturbed zebrafish embryonic haematopoiesis, with upregulated spi1 expression at the expense of gata1a. Markers associated with more differentiated myeloid cells, lcp1, lyz, and mpx were also elevated, but to a lesser extent than spi1, suggesting differentiation of early myeloid progenitors may be impaired by NUP98-HOXA9. Following irradiation, NUP98-HOXA9-expressing embryos showed increased numbers of cells in G2-M transition compared to controls and absence of a normal apoptotic response, which may result from an upregulation of bcl2. These data suggest NUP98-HOXA9-induced oncogenesis may result from a combination of defects in haematopoiesis and an aberrant response to DNA damage. Importantly, 23% of adult NUP98-HOXA9-transgenic fish developed a myeloproliferative neoplasm (MPN) at 19-23 months of age. In summary, we have identified an embryonic haematopoietic phenotype in a transgenic zebrafish line that subsequently develops MPN. This tool provides a unique opportunity for high-throughput in vivo chemical modifier screens to identify novel therapeutic agents in high risk AML.


Subject(s)
Cell Transformation, Neoplastic/genetics , Homeodomain Proteins/genetics , Leukemia, Experimental/genetics , Myeloid Cells/pathology , Myeloproliferative Disorders/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Animals , Animals, Genetically Modified , Apoptosis , Cell Cycle , Cell Lineage , DNA Damage , GATA1 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Leukemic , Genes, Reporter , Hematopoiesis/genetics , Homeodomain Proteins/physiology , Humans , Leukemia, Experimental/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Myeloid Cells/radiation effects , Myeloproliferative Disorders/pathology , Nuclear Pore Complex Proteins/physiology , Oncogene Proteins, Fusion/physiology , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Recombinant Fusion Proteins/physiology , Trans-Activators/genetics , Transgenes , Zebrafish/embryology , Zebrafish Proteins/physiology
10.
PLoS One ; 6(2): e16623, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21339820

ABSTRACT

Therapy-related acute leukemia (t-AML), is a severe complication of cytotoxic therapy used for primary cancer treatment. The outcome of these patients is poor, compared to people who develop de novo acute leukemia (p-AML). Cytogenetic abnormalities in t-AML are similar to those found in p-AML but present more frequent unfavorable karyotypes depending on the inducting agent. Losses of chromosome 5 or 7 are observed after alkylating agents while balanced translocations are found after topoisomerase II inhibitors. This study compared t-AML to p-AML using high resolution array CGH in order to find copy number abnormalities (CNA) at a higher resolution than conventional cytogenetics. More CNAs were observed in 30 t-AML than in 36 p-AML: 104 CNAs were observed with 63 losses and 41 gains (mean number 3.46 per case) in t-AML, while in p-AML, 69 CNAs were observed with 32 losses and 37 gains (mean number of 1.9 per case). In primary leukemia with a previously "normal" karyotype, 18% exhibited a previously undetected CNA, whereas in the (few) t-AML with a normal karyotype, the rate was 50%. Several minimal critical regions (MCRs) were found in t-AML and p-AML. No common MCRs were found in the two groups. In t-AML a 40 kb deleted MCR pointed to RUNX1 on 21q22, a gene coding for a transcription factor implicated in frequent rearrangements in leukemia and in familial thrombocytopenia. In de novo AML, a 1 Mb MCR harboring ERG and ETS2 was observed from patients with complex aCGH profiles. High resolution cytogenomics obtained by aCGH and similar techniques already published allowed us to characterize numerous non random chromosome abnormalities. This work supports the hypothesis that they can be classified into several categories: abnormalities common to all AML; those more frequently found in t-AML and those specifically found in p-AML.


Subject(s)
Chromosome Aberrations , Comparative Genomic Hybridization , Leukemia/genetics , Neoplasms, Second Primary/genetics , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/adverse effects , Cells, Cultured , Chromosome Aberrations/chemically induced , Comparative Genomic Hybridization/methods , DNA Copy Number Variations/physiology , Female , Gene Dosage/physiology , Humans , Karyotyping/methods , Leukemia/chemically induced , Leukemia/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Male , Middle Aged , Neoplasms, Second Primary/pathology , Young Adult
11.
Radiats Biol Radioecol ; 50(2): 195-200, 2010.
Article in Russian | MEDLINE | ID: mdl-20464968

ABSTRACT

The aim of the study was to reveal during 45 days consequences of ferromagnetic screening (FS) on the hematological and toxicogenomic indexes in rats. An express-method of nucleiod DNA content in blood by help fluorescent indication was used for alive quantitative evaluation of the toxicogenomic effects. The FS for both gender rats during 48 hrs resulted in both decreased magnetic field by 4-10 microT1 and leucopenia by 14 day of experiment. The aneu-/polyploidy index in male leucocytes was increased by 48 hr of FS and lasted out to 12-28 days after the screen removal. The leukocyte apoptosis was enhanced in female in 48 hrs only after the FS start.


Subject(s)
Electromagnetic Fields/adverse effects , Ferric Compounds , Leukemia, Radiation-Induced/etiology , Leukopenia/etiology , Animals , Apoptosis , DNA/genetics , DNA/radiation effects , Female , Leukemia, Radiation-Induced/pathology , Leukocytes/pathology , Leukocytes/radiation effects , Leukopenia/pathology , Male , Polyploidy , Rats , Time Factors
12.
Cancer Treat Res ; 145: 183-96, 2010.
Article in English | MEDLINE | ID: mdl-20306252

ABSTRACT

Primary human AML cells can be isolated and studied in vitro, but many experimental questions can only be addressed using in vivo models. In particular, tractable animal models are needed to test novel therapies. The genetic complexity of human AML poses significant challenges for the generation of reliable animal models. The hematopoietic systems of both zebrafish ( Danio rerio) and Drosophila have been well characterized ( reviewed in [5, 31]) . Both organisms are well suited to forward genetics mutagenesis screens. Although this approach has been useful for identification of mutants with hematopoietic phenotypes ( e.g., cloche), the impact on cancer biology and hematopoietic malignancies in particular has been limited. A zebrafish model of acute lymphoblastic leukemia has been generated [37] and Drosophila models have shed light on the biology of epithelial tumors ( reviewed in [60]). Nonetheless, in vivo modeling of human AML relies most heavily on mice. Most cellular, molecular, and developmental features of the hematopoietic system are well conserved across mammalian species. The availability of the human and mouse genome sequences and the capability of manipulating the mouse genome make mice the most valuable model organism for AML research. Mice have additional practical value because they have a short reproductive cycle and are relatively inexpensive to house.


Subject(s)
Disease Models, Animal , Leukemia, Myeloid, Acute , Animals , Cell Line, Tumor/transplantation , Genetic Predisposition to Disease , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Mice , Mice, Inbred NOD , Mice, Inbred Strains , Mice, SCID , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasm Transplantation , Phenotype , Retroviridae/genetics , Species Specificity , Transduction, Genetic , Transgenes , Transplantation, Heterologous
13.
J Clin Pediatr Dent ; 35(2): 203-6, 2010.
Article in English | MEDLINE | ID: mdl-21417126

ABSTRACT

UNLABELLED: Gorlin syndrome is a rare autosomal dominant disorder exhibiting high penetrance and variable expressivity. It is characterized by facial dysmorphism, skeletal anomalies, multiple basal cell carcinomas, odontogenic keratocysts (OKC), palmar and plantar pits, bifid ribs, vertebral anomalies and a variety of other malformations. Various neoplasms, such as medulloblastomas, meningiomas, ovarian and cardiac fibromas are also found in this syndrome. OBJECTIVE: To describe a twelve-year-old patient with Gorlin-Goltz syndrome, with basal cell carcinomas and promyelocytic leukemia developed after receiving craniospinal radiation for a medulloblastoma. Bifid ribs as well as mandibular and maxillar OKC were also diagnosed Conclusion: The patient with Gorlin-Goltz syndrome should receive close follow-up for early detection of malformations nd malignant neoplasias.


Subject(s)
Brain Stem Neoplasms/radiotherapy , Leukemia, Promyelocytic, Acute/pathology , Medulloblastoma/radiotherapy , Neoplasms, Multiple Primary/pathology , Neoplasms, Second Primary/pathology , Basal Cell Nevus Syndrome/pathology , Brain Stem Neoplasms/surgery , Child , Cranial Irradiation , Disease Progression , Fatal Outcome , Follow-Up Studies , Humans , Leukemia, Radiation-Induced/pathology , Male , Medulloblastoma/surgery , Neoplasm Recurrence, Local/pathology , Spine/radiation effects
14.
Br J Cancer ; 101(2): 363-71, 2009 Jul 21.
Article in English | MEDLINE | ID: mdl-19513063

ABSTRACT

BACKGROUND: The essential aetiology of radiation-induced acute myeloid leukaemia (AML) in mice is the downregulation of the transcription factor PU.1. The causative mutation of the PU.1-endocing Sfpi1 gene consists mostly of C:G to T:A transitions at a CpG site and is likely to be of spontaneous origin. To work out a mechanism underlying the association between radiation exposure and the AML induction, we have hypothesised that replicative stress after irradiation accelerates the ageing of haematopoietic stem cells (HSCs), and the ageing-related decline in DNA repair could affect the spontaneous mutation rates. METHODS: Mathematical model analysis was conducted to examine whether and to what extent the cell kinetics of HSCs can be modified after irradiation. The haematopoietic differentiation process is expressed as a mathematical model and the cell-kinetics parameters were estimated by fitting the simulation result to the assay data. RESULTS: The analysis revealed that HSCs cycle vigourously for more than a few months after irradiation. The estimated number of cell divisions per surviving HSC in 3 Gy-exposed mice reached as high as ten times that of the unexposed. INTERPRETATION: The mitotic load after 3 Gy irradiation seems to be heavy enough to accelerate the ageing of HSCs and the hypothesis reasonably explains the leukaemogenic process.


Subject(s)
Cell Division/radiation effects , Cellular Senescence/radiation effects , Hematopoietic Stem Cells/pathology , Leukemia, Myeloid/pathology , Leukemia, Radiation-Induced/pathology , Acute Disease , Animals , Cell Division/physiology , Cellular Senescence/physiology , Leukemia, Myeloid/etiology , Male , Mice , Mice, Inbred C3H , Mitosis/physiology , Mitosis/radiation effects , Models, Biological
15.
Eur J Med Res ; 14(1): 42-3, 2009 Jan 28.
Article in English | MEDLINE | ID: mdl-19258210

ABSTRACT

The occurrence of acute promyelocytic leukemia (APL) in HIV-infected patients has been reported in only five cases. Due to a very small number of reported HIV/APL patients who have been treated with different therapies with the variable outcome, the prognosis of APL in the setting of the HIV-infection is unclear. Here, we report a case of an HIV-patient who developed APL and upon treatment entered a complete remission. A 25-years old male patient was diagnosed with HIV-infection in 1996, but remained untreated. In 2004, the patient was diagnosed with primary central nervous system lymphoma. We treated the patient with antiretroviral therapy and whole-brain irradiation, resulting in complete remission of the lymphoma. In 2006, prompted by a sudden neutropenia, we carried out a set of diagnostic procedures, revealing APL. Induction therapy consisted of standard treatment with all-trans-retinoic-acid (ATRA) and idarubicin. Subsequent cytological and molecular ana?lysis of bone marrow demonstrated complete hematological and molecular remission. Due to the poor general condition, consolidation treatment with ATRA was given in March and April 2007. The last follow-up 14 months later, showed sustained molecular APL remission. In conclusion, we demonstrated that a complete molecular APL remission in an HIV-patient was achieved by using reduced-intensity treatment.


Subject(s)
Brain Neoplasms/radiotherapy , Brain/radiation effects , HIV Infections/complications , Leukemia, Promyelocytic, Acute/etiology , Leukemia, Radiation-Induced/etiology , Lymphoma/radiotherapy , Adult , Anti-Retroviral Agents/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Antiretroviral Therapy, Highly Active/methods , Bisexuality , HIV Infections/drug therapy , Humans , Idarubicin/therapeutic use , Leukemia, Promyelocytic, Acute/pathology , Leukemia, Radiation-Induced/pathology , Male , Remission Induction , Tretinoin
16.
J Pathol ; 214(5): 610-6, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18266203

ABSTRACT

The major adverse consequences of radiation exposure, including the initiation of leukaemia and other malignancies, are generally attributed to effects in the cell nucleus at the time of irradiation. However, genomic damage as a longer term consequence of radiation exposure has more recently been demonstrated due to untargeted radiation effects including delayed chromosomal instability and bystander effects. These processes, mainly studied in vitro, are characterized by un-irradiated cells demonstrating effects as though they themselves had been irradiated and have been associated with altered oxidative processes. To investigate the potential for these untargeted effects of radiation to produce delayed damaging events in vivo, we studied a well-characterized model of radiation-induced acute myeloid leukaemia in CBA/Ca mice. Haemopoietic tissues of irradiated CBA/Ca mice exhibit enhanced levels of p53 stabilization, increased levels of p21(waf1), and increased amounts of apoptosis, as expected, in the first few hours post-irradiation, but also at much later times: weeks and months after the initial exposure. Because these responses are seen in cells that were not themselves directly irradiated but are the descendants of irradiated cells, the data are consistent with an initial radiation exposure leading to persistently increased levels of ongoing DNA damage, analogous to radiation-induced chromosomal instability. To investigate the potential source of ongoing oxidative processes, we show increased levels of 3-nitrotyrosine, a marker of damaging nitrogen/oxygen species in macrophages. Not all animals show increased oxidative activity or p53 responses as long-term consequences of irradiation, but increased levels of p53, p21, and apoptosis are directly correlated with increased 3-nitrotyrosine in individual mice post-irradiation. The data implicate persistent activation of inflammatory-type responses in irradiated tissues as a contributory bystander mechanism for causing delayed DNA damage.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Radiation-Induced/metabolism , Macrophages/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Bystander Effect , DNA Damage , DNA, Neoplasm/genetics , Immunoenzyme Techniques , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Leukemia, Radiation-Induced/genetics , Leukemia, Radiation-Induced/pathology , Mice , Mice, Inbred CBA , Signal Transduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Leukemia ; 21(12): 2506-11, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17805326

ABSTRACT

Previous studies have shown that high levels of MMP-9 can be detected in the serum of patients with various lymphoid malignancies and in leukemia/lymphoma culture supernatants. Indeed, aggressive forms of lymphoma constitutively produce MMP-9 and its elevated levels in the serum or in tissues correlate with advanced stage and poor patient survival. In vitro, MMP-9, which is also produced by the host peritumoral cells in response to the presence of tumors, plays an important role in migration of tumor cells through artificial basement membranes or endothelial cells. In this study, using MMP-9-deficient mice, we show that absence of MMP-9 does not prevent the development of primary T-cell leukemia. Furthermore, MMP-9-deficient cell lines retained their tumorigenic potential, as shown by their ability to induce thymic lymphoma in young syngeneic wild-type animals. In addition, these MMP-9-deficient tumor cells disseminate in normal mice, or mice that are deficient for MMP-9, indicating that tumor growth and dissemination can occur in total absence of MMP-9. These results show for the first time than lymphoma growth can occur in total absence of MMP-9 and have consequences for therapy of invasive cancers with inhibitors of MMPs.


Subject(s)
Leukemia, Radiation-Induced/enzymology , Leukemia, T-Cell/enzymology , Lymphoma, T-Cell/enzymology , Matrix Metalloproteinase 9/deficiency , Neoplasm Proteins/deficiency , Neoplasms, Radiation-Induced/enzymology , Thymus Neoplasms/enzymology , Animals , Cell Line, Tumor/transplantation , Crosses, Genetic , Female , Leukemia, Radiation-Induced/pathology , Leukemia, T-Cell/etiology , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/pathology , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Neoplasms, Radiation-Induced/pathology , Specific Pathogen-Free Organisms , Thymus Neoplasms/etiology , Thymus Neoplasms/pathology
18.
Exp Hematol ; 35(4 Suppl 1): 125-33, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17379097

ABSTRACT

OBJECTIVE: The aim of this study is to investigate a possible implication in cell kinetics of the hematopoietic progenitors to the experimental leukemogenesis to elucidate the relevance of various leukemic mode of action to Gompertzean survival curves, a new parameter based on the lifespan. MATERIALS AND METHODS: Mice, C3H/He, and C57BL/6 strain, male and female, with or without genetic modifications, e.g., p53-deficiency or thioredoxin overexpression were used in the present hemopoietic stem/progenitor research, radiation- or benzene-induced leukemogenesis followed by histopathological examination. A lethal dose of radiation for bone marrow transplantation, and a graded increased dose up to 5 Gy of x-rays for induction of hematopoietic malignancies were given. For caloric restriction studies, 77 kcal/week was maintained in accordance to different restriction-timing. For assays of hematopoietic colonization, colony-forming unit spleen and colony-forming unit granulocyte macrophage were evaluated. Hematopoietic progenitor cell-specific kinetics were studied by continuous labeling of bromodeoxyuridine for cycling cells, followed by ultraviolet (UV) exposure and hemopoietic colonization (bromodeoxyuridine UV [BUUV] method). Various experimental survival curves were applied to a mathematical analysis by Gompertz-Makeham law of mortality. RESULTS: Referring current authors' studies on leukemogenesis induced by ionizing radiation and benzene exposure, implications of hematopoietic progenitor cell kinetics to the experimental leukemogenesis were evaluated by means of a novel experimental tool, the BUUV method. Comparative studies to elucidate relevancies of these data, including two prevention studies, one on caloric restriction and the other on antioxidative thioredoxin overexpression, to those Gompertzean survival curves of experimental animals were analyzed. CONCLUSION: The Gompertzean expression may elucidate an appropriate toxicological endpoint for evaluating the effect of radiation and/or benzene-exposure on the lifespan and its modification by various experimental preventive measures.


Subject(s)
Benzene/toxicity , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Leukemia, Radiation-Induced/metabolism , Models, Biological , Animals , Bone Marrow Transplantation , Caloric Restriction , Disease Models, Animal , Dose-Response Relationship, Radiation , Endpoint Determination , Female , Hematopoietic Stem Cells/pathology , Humans , Kinetics , Leukemia, Radiation-Induced/mortality , Leukemia, Radiation-Induced/pathology , Leukemia, Radiation-Induced/prevention & control , Male , Mice , Tumor Stem Cell Assay
19.
J Radiat Res ; 47(2): 121-30, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16819138

ABSTRACT

The aim is to clarify expression changes of ERK1/2, STAT3 and SHP-2 in bone marrow cells from gamma-ray induced leukemia mice. A mouse model of gamma-ray induced leukemia was produced, and by means of quantitative real-time PCR, immunoprecipitation, Western blotting and electrophoretic mobility shift assays (EMSA), the expression of mRNA and protein, phosphorylation level, and protein activity of ERK1/2, STAT3 and SHP-2 in bone marrow cells were investigated in these mice. The results indicated that mRNA and protein expressions of ERK1/2 were upregulated, with significant increase of phosphorylation level and protein activity, but with insignificant differences in mRNA and protein expressions, phosphorylation level and protein activity of STAT3 and SHP-2 in bone marrow cells from gamma-ray induced leukemia mice compared to the radiation/tumor-free or control mice. It is concluded that in the pathogenesis of gamma-ray induced leukemia in Balb/C mice, activated ERK1/2 pathway may play a role, without involving STAT3 pathway; meanwhile, SHP-2 exerts no regulative effect on pathways of Ras-ERK1/2 and JAK-STAT.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/radiation effects , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Radiation-Induced/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Protein Tyrosine Phosphatases/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Female , Gamma Rays/adverse effects , Gene Expression Regulation, Neoplastic/radiation effects , Leukemia, Radiation-Induced/etiology , Leukemia, Radiation-Induced/pathology , Male , Mice , Mice, Inbred BALB C , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Whole-Body Irradiation/adverse effects
20.
J Radiat Res ; 45(2): 245-51, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15304967

ABSTRACT

Allelic loss on the chromosome 2 is associated with radiation-induced murine acute myeloid leukemia. However, the gene, which contributes mainly to the leukemogenesis has not yet been identified. Expecting any predisposition to acute myeloid leukemia, we performed a radiation leukemogenensis experiment with Pax6(Sey3H), one of the small eye mutants carrying a congenital hemizygosity of the chromosome 2 middle region. A deletion mapping of Pax6(Sey3H) with 50 STS markers indicated that the deleted segment extended between the 106.00 and 111.47 Mb site from the centromere with a length of 5.47 Mb. In the deleted segment, 6 known and 17 novel genes were located. Pax6(Sey3H) mutants that crossed back into C3H/He did not develop myeloid leukemia spontaneously, but they did when exposed to gamma-rays. The final incidence of myeloid leukemia in mutants (25.8%) was as high as that in normal sibs (21.4%). Survival curves of leukemia-bearing mutants shifted toward the left (p = 0.043 by the Log rank test). F1 hybrids of Pax6(Sey3H) with JF1 were less susceptible to radiation than Pax6(Sey3H) onto C3H/He in regard to survival (p = 0.003 and p < 0.00001 for mutants and normal sibs, respectively, by a test of the difference between two proportions). Congenital deletion of the 5.47 Mb segment at the middle region on chromosome 2 alone did not trigger myeloid stem cells to expand clonally in vivo; however, the deletion shortcut the latency of radiation-induced myeloid leukemia.


Subject(s)
Aniridia/pathology , Genetic Predisposition to Disease/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intestinal Neoplasms/pathology , Leukemia, Myeloid, Acute/pathology , Leukemia, Radiation-Induced/pathology , Animals , Aniridia/complications , Eye Proteins , Gamma Rays , Intestinal Neoplasms/complications , Leukemia, Myeloid, Acute/complications , Leukemia, Radiation-Induced/complications , Mice , Mice, Inbred C3H , Mice, Knockout , PAX6 Transcription Factor , Paired Box Transcription Factors , Repressor Proteins , Survival , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...