1
|
Karabulutoglu M, Finnon R, Cruz-Garcia L, Hill MA, Badie C. Oxidative Stress and X-ray Exposure Levels-Dependent Survival and Metabolic Changes in Murine HSPCs. Antioxidants (Basel) 2021; 11:11. [PMID: 35052515 PMCID: PMC8772903 DOI: 10.3390/antiox11010011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/10/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Haematopoietic bone marrow cells are amongst the most sensitive to ionizing radiation (IR), initially resulting in cell death or genotoxicity that may later lead to leukaemia development, most frequently Acute Myeloid Leukaemia (AML). The target cells for radiation-induced Acute Myeloid Leukaemia (rAML) are believed to lie in the haematopoietic stem and progenitor cell (HSPC) compartment. Using the inbred strain CBA/Ca as a murine model of rAML, progress has been made in understanding the underlying mechanisms, characterisation of target cell population and responses to IR. Complex regulatory systems maintain haematopoietic homeostasis which may act to modulate the risk of rAML. However, little is currently known about the role of metabolic factors and diet in these regulatory systems and modification of the risk of AML development. This study characterises cellular proliferative and clonogenic potential as well as metabolic changes within murine HSPCs under oxidative stress and X-ray exposure. Ambient oxygen (normoxia; 20.8% O2) levels were found to increase irradiated HSPC-stress, stimulating proliferative activity compared to low oxygen (3% O2) levels. IR exposure has a negative influence on the proliferative capability of HSPCs in a dose-dependent manner (0-2 Gy) and this is more pronounced under a normoxic state. One Gy x-irradiated HSPCs cultured under normoxic conditions displayed a significant increase in oxygen consumption compared to those cultured under low O2 conditions and to unirradiated HSPCs. Furthermore, mitochondrial analyses revealed a significant increase in mitochondrial DNA (mtDNA) content, mitochondrial mass and membrane potential in a dose-dependent manner under normoxic conditions. Our results demonstrate that both IR and normoxia act as stressors for HSPCs, leading to significant metabolic deregulation and mitochondrial dysfunctionality which may affect long term risks such as leukaemia.
Collapse
Affiliation(s)
- Melis Karabulutoglu
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Radiation, Chemical and Environmental Hazards Directorate (RCE, Formally CRCE), UK Health Security Agency (Formerly Public Health England), Chilton, Didcot, Oxon OX11 0RQ, UK; (R.F.); (L.C.-G.)
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK;
| | - Rosemary Finnon
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Radiation, Chemical and Environmental Hazards Directorate (RCE, Formally CRCE), UK Health Security Agency (Formerly Public Health England), Chilton, Didcot, Oxon OX11 0RQ, UK; (R.F.); (L.C.-G.)
| | - Lourdes Cruz-Garcia
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Radiation, Chemical and Environmental Hazards Directorate (RCE, Formally CRCE), UK Health Security Agency (Formerly Public Health England), Chilton, Didcot, Oxon OX11 0RQ, UK; (R.F.); (L.C.-G.)
| | - Mark A. Hill
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK;
| | - Christophe Badie
- Cancer Mechanisms and Biomarkers Group, Radiation Effects Department, Radiation, Chemical and Environmental Hazards Directorate (RCE, Formally CRCE), UK Health Security Agency (Formerly Public Health England), Chilton, Didcot, Oxon OX11 0RQ, UK; (R.F.); (L.C.-G.)
| |
Collapse
|
2
|
Frosina G, Fontana V, Verzola D, Rosa A, Gaggero G, Garibotto G, Vagge S, Pigozzi S, Daga A. Ultra-hyper-fractionated radiotherapy for high-grade gliomas. J Neurosci Res 2021; 99:3182-3203. [PMID: 34747065 DOI: 10.1002/jnr.24929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
High-grade gliomas (HGGs; WHO grades III and IV) are invariably lethal brain tumors. Low-dose hyper-radiosensitivity (HRS) of HGG is a well-established phenomenon in vitro. However, possibly linked to the unavailability of accurate animal models of the diseases, this therapeutic effect could not be consistently translated to the animal setting, thus impairing its subsequent clinical development. The purpose of this study was to develop radiotherapeutic (RT) schedules permitting to significantly improve the overall survival of faithful animal models of HGG that have been recently made available. We used primary glioma initiating cell (GIC)-driven orthotopic animal models that accurately recapitulate the heterogeneity and growth patterns of the patients' tumors, to investigate the therapeutic effects of low radiation doses toward HGG. With the same total dose, RT fractions ≤0.5 Gy twice per week [ultra-hyper-fractionation (ultra-hyper-FRT)] started at early stages of tumor progression (a condition that in the clinical setting often occurs at the end of the guidelines treatment) improved the effectiveness of RT and the animal survival in comparison to standard fractions. For the same cumulative dose, the use of fractions ≤0.5 Gy may permit to escape one or more tumor resistance mechanisms thus increasing the effectiveness of RT and the overall animal survival. These findings suggest investigating in the clinical setting the therapeutic effect of an ultra-hyper-FRT schedule promptly extending the conventional RT component of the current guideline ("Stupp") therapeutic protocol.
Collapse
Affiliation(s)
- Guido Frosina
- Mutagenesis & Cancer Prevention, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Vincenzo Fontana
- Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Department of Internal Medicine and Medical Specialties - Dimi, University of Genova, Genova, Italy
| | - Alessandra Rosa
- Clinical Epidemiology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gabriele Gaggero
- Pathological Anatomy and Histology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giacomo Garibotto
- Department of Internal Medicine and Medical Specialties - Dimi, University of Genova, Genova, Italy
| | - Stefano Vagge
- Radiation Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Simona Pigozzi
- Department of Surgical Sciences and Integrated Diagnostics - Disc, University of Genova, Genova, Italy
| | - Antonio Daga
- Cellular Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
3
|
Etani R, Ojima M, Ariyoshi K, Fujishima Y, Kai M. Cellular kinetics of hematopoietic cells with Sfpi1 deletion are present at different frequencies in bone-marrow and spleen in X-irradiated mice. Int J Radiat Biol 2020; 96:1119-1124. [PMID: 32658559 DOI: 10.1080/09553002.2020.1793018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Several past studies using a mouse model of radiation-induced AML (rAML) have shown that hemizygous deletion of the Sfpi1 gene (HDSG) is an initiating event for the development of rAML. In this study, we examined the difference in frequency of HDSG in hematopoietic stem cells (HSCs) Rich hematopoietic Cell population (HRCs) from bone marrow (BM) and spleen of C3H mice irradiated with 3 Gy X-rays. MATERIALS AND METHODS 8-weeks old male C3H mice were irradiated 3Gy of whole body X-ray (1 Gy/min) and mice were sacrificed at 1, 4, 8, and 26 weeks. Then, HSPCs were isolated from BM of femur and spleen, the frequency of HRCs with Sfpi1 gene deletion was analyzed by fluorescence in situ hybridization (FISH). RESULTS AND CONCLUSIONS The frequency of HRCs with HDSG in both BM and spleen was increased 1 week after X-irradiation. Then, the frequency of HRCs with HDSG in BM showed a gradual decrease from 4 to 26 weeks, whereas HRCs with HDSG in spleen remained high, even at 26 weeks after X-irradiation. HDSG is less likely to be eliminated, particularly in the spleen, after X-irradiation. The spleen as well as BM of the femur may be major sites of rAML development.
Collapse
Affiliation(s)
- Reo Etani
- Laboratory of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita City, Japan
| | - Mitsuaki Ojima
- Laboratory of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita City, Japan
| | - Kentaro Ariyoshi
- Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki City, Japan.,Integrated Center for Science and Humanities, Fukushima Medical University, Fukushima City, Japan
| | - Yohei Fujishima
- Institute of Radiation Emergency Medicine, Hirosaki University, Hirosaki City, Japan.,Department of Radiation Biology, Tohoku University Grauduate School of Medicine, Sendai City, Japan
| | - Michiaki Kai
- Laboratory of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita City, Japan
| |
Collapse
|
4
|
Ojima M, Hirouchi T, Etani R, Ariyoshi K, Fujishima Y, Kai M. Dose-Rate-Dependent PU.1 Inactivation to Develop Acute Myeloid Leukemia in Mice Through Persistent Stem Cell Proliferation After Acute or Chronic Gamma Irradiation. Radiat Res 2019; 192:612-620. [PMID: 31560640 DOI: 10.1667/rr15359.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced acute myeloid leukemia (rAML) in C3H mice is commonly developed through inactivation of PU.1 transcription factor encoded in Sfpi1 on chromosome 2. PU.1 inactivation involves two steps: hemizygous deletion of the Sfpi1 gene (DSG) and point mutation of the allele Sfpi1 gene (PMASG). In this study, we investigated the dose-rate dependence of the frequency of both DSG and PMASG in hematopoietic stem cells (HSCs) of C3H mice that received a total of 3 Gy gamma-ray exposure at dose rates of 20 mGy/day, 200 mGy/day or 1,000 mGy/min. All mice were followed for 250 days from start of irradiation. Fluorescent in situ hybridization of the Sfpi1 gene site indicated that frequency of HSCs with DSG was proportional to dose rate. In cell surface profiles, PU.1-inactivated HSCs by both DSG and PMASG were still positive for PU.1, but negative for GM-CSF receptor-α (GMCSFRα), which is transcriptionally regulated by PU.1. Immunofluorescent staining analysis of both PU.1 and GM-CSFRα also showed dose-rate-dependent levels of PU.1-inactivated HSCs. This study provides evidence that both DSG and PMASG are dose-rate dependent; these experimental data offer new insights into the dose-rate effects in HSCs that can lead to radiation-induced leukemogenesis.
Collapse
Affiliation(s)
- Mitsuaki Ojima
- Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | | | - Reo Etani
- Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| | - Kentaro Ariyoshi
- Institute of Radiation Emergency Medicine, Hirosaki University, Aomori 036-8564, Japan and Department of Radiation Biology, Tohoku University School of Medicine, Sendai, Japan
| | - Yohei Fujishima
- Institute of Radiation Emergency Medicine, Hirosaki University, Aomori 036-8564, Japan and Department of Radiation Biology, Tohoku University School of Medicine, Sendai, Japan
| | - Michiaki Kai
- Department of Environmental Health Science, Oita University of Nursing and Health Sciences, Oita 840-1201, Japan
| |
Collapse
|
5
|
Gault N, Verbiest T, Badie C, Romeo PH, Bouffler S. Hematopoietic stem and progenitor cell responses to low radiation doses - implications for leukemia risk. Int J Radiat Biol 2019; 95:892-899. [PMID: 30652952 DOI: 10.1080/09553002.2019.1569777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Studies of the responses of hematopoietic stem and progenitor cells (HSPCs) to low doses of ionizing radiation formed an important aspect of the RISK-IR project ( www.risk-ir.eu ). A brief overview of these studies is presented here. The findings confirm the sensitivity of HSPCs to radiation even at low doses, and illustrate the substantial impact that differentiation state has upon cell sensitivity. The work provides mechanistic support for epidemiological findings of leukemia risk at dose levels used in diagnostic CT imaging, and further suggests that low-dose irradiation may facilitate bone marrow transplantation, a finding that could lead to refinements in clinical practice.
Collapse
Affiliation(s)
- Nathalie Gault
- a CEA/DRF/IBFJ/iRCM/LRTS , Fontenay-aux-Roses Cedex , France.,b Inserm U967 , Fontenay-aux-Roses Cedex , France.,c CEA/DRF/IBFJ/iRCM/LRTS-U1274 Inserm-Université Paris-Diderot , Paris , France.,d CEA/DRF/IBFJ/iRCM/LRTS-U1274 Inserm-Université Paris-Sud , Paris , France
| | - Tom Verbiest
- e Public Health England , Centre for Radiation, Chemical and Environmental Hazards , Oxfordshire , UK
| | - Christophe Badie
- e Public Health England , Centre for Radiation, Chemical and Environmental Hazards , Oxfordshire , UK
| | - Paul-Henri Romeo
- a CEA/DRF/IBFJ/iRCM/LRTS , Fontenay-aux-Roses Cedex , France.,b Inserm U967 , Fontenay-aux-Roses Cedex , France.,c CEA/DRF/IBFJ/iRCM/LRTS-U1274 Inserm-Université Paris-Diderot , Paris , France.,d CEA/DRF/IBFJ/iRCM/LRTS-U1274 Inserm-Université Paris-Sud , Paris , France
| | - Simon Bouffler
- e Public Health England , Centre for Radiation, Chemical and Environmental Hazards , Oxfordshire , UK
| |
Collapse
|
6
|
Shimura N, Kojima S. The Lowest Radiation Dose Having Molecular Changes in the Living Body. Dose Response 2018; 16:1559325818777326. [PMID: 29977175 PMCID: PMC6024299 DOI: 10.1177/1559325818777326] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 12/20/2022] Open
Abstract
We herein attempted to identify the lowest radiation dose causing molecular changes in the living body. We investigated the effects of radiation in human cells, animals, and humans. DNA double-strand breaks (DSBs) formed in cells at γ- or X-ray irradiation doses between 1 mGy and 0.5 Gy; however, the extent of DSB formation differed depending on the cell species. The formation of micronuclei (MNs) and nucleoplasmic bridges (NPBs) was noted at radiation doses between 0.1 and 0.2 Gy. Stress-responsive genes were upregulated by lower radiation doses than those that induced DNA DSBs or MN and NPBs. These γ- or X-ray radiation doses ranged between approximately 10 and 50 mGy. In animals, chromosomal aberrations were detected between 50 mGy and 0.1 Gy of low linear energy transfer radiation, 0.1 Gy of metal ion beams, and 9 mGy of fast neutrons. In humans, DNA damage has been observed in children who underwent computed tomography scans with an estimated blood radiation dose as low as 0.15 mGy shortly after examination. The frequencies of chromosomal translocations were lower in residents of high background areas than in those of control areas. In humans, systemic adaptive responses may have been prominently expressed at these radiation doses.
Collapse
Affiliation(s)
- Noriko Shimura
- Faculty of Pharmaceutical Sciences, Ohu University, Tomita-machi, Koriyama, Fukushima, Japan
| | - Shuji Kojima
- Faculty of Pharmaceutical Sciences, Department of Radiation Biosciences, Tokyo University of Science (TUS), Chiba, Japan
| |
Collapse
|
7
|
Patel R, Qing Y, Kennedy L, Yan Y, Pink J, Aguila B, Desai A, Gerson SL, Welford SM. MMR Deficiency Does Not Sensitize or Compromise the Function of Hematopoietic Stem Cells to Low and High LET Radiation. Stem Cells Transl Med 2018; 7:513-520. [PMID: 29656536 PMCID: PMC6052615 DOI: 10.1002/sctm.17-0295] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/20/2018] [Indexed: 12/12/2022] Open
Abstract
One of the major health concerns on long-duration space missions will be radiation exposure to the astronauts. Outside the earth's magnetosphere, astronauts will be exposed to galactic cosmic rays (GCR) and solar particle events that are principally composed of protons and He, Ca, O, Ne, Si, Ca, and Fe nuclei. Protons are by far the most common species, but the higher atomic number particles are thought to be more damaging to biological systems. Evaluation and amelioration of risks from GCR exposure will be important for deep space travel. The hematopoietic system is one of the most radiation-sensitive organ systems, and is highly dependent on functional DNA repair pathways for survival. Recent results from our group have demonstrated an acquired deficiency in mismatch repair (MMR) in human hematopoietic stem cells (HSCs) with age due to functional loss of the MLH1 protein, suggesting an additional risk to astronauts who may have significant numbers of MMR deficient HSCs at the time of space travel. In the present study, we investigated the effects gamma radiation, proton radiation, and 56 Fe radiation on HSC function in Mlh1+/+ and Mlh1-/- marrow from mice in a variety of assays and have determined that while cosmic radiation is a major risk to the hematopoietic system, there is no dependence on MMR capacity. Stem Cells Translational Medicine 2018;7:513-520.
Collapse
Affiliation(s)
| | - Yulan Qing
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Lucy Kennedy
- Unit for Laboratory and Animal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yan Yan
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - John Pink
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Brittany Aguila
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - Amar Desai
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Stanton L Gerson
- Case Comprehensive Cancer Center, National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical center and Case Western Reserve University, Cleveland, Ohio, USA
| | - Scott M Welford
- Department of Radiation Oncology, Sylvester Cancer Center, University of Miami, Miami, Florida, USA
| |
Collapse
|
8
|
Tracking preleukemic cells in vivo to reveal the sequence of molecular events in radiation leukemogenesis. Leukemia 2018; 32:1435-1444. [PMID: 29556020 PMCID: PMC5990525 DOI: 10.1038/s41375-018-0085-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/05/2018] [Accepted: 02/06/2018] [Indexed: 12/16/2022]
Abstract
Epidemiological studies have demonstrated an increased leukemia incidence following ionizing radiation exposure, but to date, the target cells and underlying mechanisms of radiation leukemogenesis remain largely unidentified. We engineered a mouse model carrying a different fluorescent marker on each chromosome 2, located inside the minimum deleted region occurring after radiation exposure and recognized as the first leukemogenic event. Using this tailored model, we report that following radiation exposure, more than half of asymptomatic CBA Sfpi1GFP/mCh mice presented with expanding clones of preleukemic hematopoietic cells harboring a hemizygous interstitial deletion of chromosome 2. Moreover, following isolation of preleukemic hematopoietic stem and progenitor cells irradiated in their native microenvironment, we identified the presence of Sfpi1 point mutations within a subpopulation of these preleukemic cells expanding rapidly (increasing from 6% to 55% in 21 days in peripheral blood in one case), hence identifying for the first time the presence of such cells within a living animal. Importantly, we also report a previously undescribed gender difference in the phenotype of the preleukemic cells and leukemia, suggesting a gender imbalance in the radiation-induced leukemic target cell. In conclusion, we provide novel insights into the sequence of molecular events occurring during the (radiation-induced) leukemic clonal evolution.
Collapse
|
9
|
In vitro and in vivo assessment of direct effects of simulated solar and galactic cosmic radiation on human hematopoietic stem/progenitor cells. Leukemia 2016; 31:1398-1407. [PMID: 27881872 DOI: 10.1038/leu.2016.344] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 10/10/2016] [Accepted: 10/21/2016] [Indexed: 12/13/2022]
Abstract
Future deep space missions to Mars and near-Earth asteroids will expose astronauts to chronic solar energetic particles (SEP) and galactic cosmic ray (GCR) radiation, and likely one or more solar particle events (SPEs). Given the inherent radiosensitivity of hematopoietic cells and short latency period of leukemias, space radiation-induced hematopoietic damage poses a particular threat to astronauts on extended missions. We show that exposing human hematopoietic stem/progenitor cells (HSC) to extended mission-relevant doses of accelerated high-energy protons and iron ions leads to the following: (1) introduces mutations that are frequently located within genes involved in hematopoiesis and are distinct from those induced by γ-radiation; (2) markedly reduces in vitro colony formation; (3) markedly alters engraftment and lineage commitment in vivo; and (4) leads to the development, in vivo, of what appears to be T-ALL. Sequential exposure to protons and iron ions (as typically occurs in deep space) proved far more deleterious to HSC genome integrity and function than either particle species alone. Our results represent a critical step for more accurately estimating risks to the human hematopoietic system from space radiation, identifying and better defining molecular mechanisms by which space radiation impairs hematopoiesis and induces leukemogenesis, as well as for developing appropriately targeted countermeasures.
Collapse
|
10
|
Influence of radiation quality on mouse chromosome 2 deletions in radiation-induced acute myeloid leukaemia. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 793:48-54. [DOI: 10.1016/j.mrgentox.2015.07.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 01/21/2023]
|
11
|
Bacher JW, Sievers CK, Albrecht DM, Grimes IC, Weiss JM, Matkowskyj KA, Agni RM, Vyazunova I, Clipson L, Storts DR, Thliveris AT, Halberg RB. Improved Detection of Microsatellite Instability in Early Colorectal Lesions. PLoS One 2015; 10:e0132727. [PMID: 26252492 PMCID: PMC4529134 DOI: 10.1371/journal.pone.0132727] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/17/2015] [Indexed: 12/22/2022] Open
Abstract
Microsatellite instability (MSI) occurs in over 90% of Lynch syndrome cancers and is considered a hallmark of the disease. MSI is an early event in colon tumor development, but screening polyps for MSI remains controversial because of reduced sensitivity compared to more advanced neoplasms. To increase sensitivity, we investigated the use of a novel type of marker consisting of long mononucleotide repeat (LMR) tracts. Adenomas from 160 patients, ranging in age from 29–55 years old, were screened for MSI using the new markers and compared with current marker panels and immunohistochemistry standards. Overall, 15 tumors were scored as MSI-High using the LMRs compared to 9 for the NCI panel and 8 for the MSI Analysis System (Promega). This difference represents at least a 1.7-fold increase in detection of MSI-High lesions over currently available markers. Moreover, the number of MSI-positive markers per sample and the size of allelic changes were significantly greater with the LMRs (p = 0.001), which increased confidence in MSI classification. The overall sensitivity and specificity of the LMR panel for detection of mismatch repair deficient lesions were 100% and 96%, respectively. In comparison, the sensitivity and specificity of the MSI Analysis System were 67% and 100%; and for the NCI panel, 75% and 97%. The difference in sensitivity between the LMR panel and the other panels was statistically significant (p<0.001). The increased sensitivity for detection of MSI-High phenotype in early colorectal lesions with the new LMR markers indicates that MSI screening for the early detection of Lynch syndrome might be feasible.
Collapse
Affiliation(s)
- Jeffery W. Bacher
- Genetic Analysis Group, Promega Corporation, Madison, Wisconsin, United States of America
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Chelsie K. Sievers
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Dawn M. Albrecht
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Ian C. Grimes
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Jennifer M. Weiss
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Kristina A. Matkowskyj
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Rashmi M. Agni
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Irina Vyazunova
- Genetic Analysis Group, Promega Corporation, Madison, Wisconsin, United States of America
| | - Linda Clipson
- Department of Oncology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Douglas R. Storts
- Genetic Analysis Group, Promega Corporation, Madison, Wisconsin, United States of America
| | - Andrew T. Thliveris
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Richard B. Halberg
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
12
|
Verbiest T, Bouffler S, Nutt SL, Badie C. PU.1 downregulation in murine radiation-induced acute myeloid leukaemia (AML): from molecular mechanism to human AML. Carcinogenesis 2015; 36:413-9. [PMID: 25750172 PMCID: PMC4392607 DOI: 10.1093/carcin/bgv016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/24/2015] [Indexed: 01/06/2023] Open
Abstract
The transcription factor PU.1, encoded by the murine Sfpi1 gene (SPI1 in humans), is a member of the Ets transcription factor family and plays a vital role in commitment and maturation of the myeloid and lymphoid lineages. Murine studies directly link primary acute myeloid leukaemia (AML) and decreased PU.1 expression in specifically modified strains. Similarly, a radiation-induced chromosome 2 deletion and subsequent Sfpi1 point mutation in the remaining allele lead to murine radiation-induced AML. Consistent with murine data, heterozygous deletion of the SPI1 locus and mutation of the −14kb SPI1 upstream regulatory element were described previously in human primary AML, although they are rare events. Other mechanisms linked to PU.1 downregulation in human AML include TP53 deletion, FLT3-ITD mutation and the recurrent AML1-ETO [t(8;21)] and PML-RARA [t(15;17)] translocations. This review provides an up-to-date overview on our current understanding of the involvement of PU.1 in the initiation and development of radiation-induced AML, together with recommendations for future murine and human studies.
Collapse
Affiliation(s)
- Tom Verbiest
- Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK, CRUK & MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Simon Bouffler
- Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christophe Badie
- Biological Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Didcot OX11 ORQ, UK,
| |
Collapse
|
13
|
Genik PC, Vyazunova I, Steffen LS, Bacher JW, Bielefeldt-Ohmann H, McKercher S, Ullrich RL, Fallgren CM, Weil MM, Ray FA. Leukemogenesis in heterozygous PU.1 knockout mice. Radiat Res 2014; 182:310-5. [PMID: 25076114 DOI: 10.1667/rr13738.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Most murine radiation-induced acute myeloid leukemias involve biallelic inactivation of the PU.1 gene, with one allele being lost through a radiation-induced chromosomal deletion and the other allele affected by a recurrent point mutation in codon 235 that is likely to be spontaneous. The short latencies of acute myeloid leukemias occurring in nonirradiated mice engineered with PU.1 conditional knockout or knockdown alleles suggest that once both copies of PU.1 have been lost any other steps involved in leukemogenesis occur rapidly. Yet, spontaneous acute myeloid leukemias have not been reported in mice heterozygous for a PU.1 knockout allele, an observation that conflicts with the understanding that the PU.1 codon 235 mutation is spontaneous. Here we describe experiments that show that the lack of spontaneous leukemia in PU.1 heterozygous knockout mice is not due to insufficient monitoring times or mouse numbers or the genetic background of the knockout mice. The results reveal that spontaneous leukemias that develop in mice of the mixed 129S2/SvPas and C57BL/6 background of knockout mice arise by a pathway that does not involve biallelic PU.1 mutation. In addition, the latency of radiation-induced leukemia in PU.1 heterozygous mice on a genetic background susceptible to radiation-induced leukemia indicates that the codon 235 mutation is not a rate-limiting step in radiation leukemogenesis driven by PU.1 loss.
Collapse
Affiliation(s)
- Paula C Genik
- a Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Miousse IR, Shao L, Chang J, Feng W, Wang Y, Allen AR, Turner J, Stewart B, Raber J, Zhou D, Koturbash I. Exposure to low-dose (56)Fe-ion radiation induces long-term epigenetic alterations in mouse bone marrow hematopoietic progenitor and stem cells. Radiat Res 2014; 182:92-101. [PMID: 24960414 DOI: 10.1667/rr13580.1] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There is an increasing need to better understand the long-term health effects of high-linear energy transfer (LET) radiation due to exposure during space missions, as well as its increasing use in clinical treatments. Previous studies have indicated that exposure to (56)Fe heavy ions increases the incidence of acute myeloid leukemia (AML) in mice but the underlying molecular mechanisms remain elusive. Epigenetic alterations play a role in radiation-induced genomic instability and the initiation and progression of AML. In this study, we assessed the effects of low-dose (56)Fe-ion irradiation on epigenetic alterations in bone marrow mononuclear cells (BM-MNCs) and hematopoietic progenitor and stem cells (HPSCs). Exposure to (56)Fe ions (600 MeV, 0.1, 0.2 and 0.4 Gy) resulted in significant epigenetic alterations involving methylation of DNA, the DNA methylation machinery and expression of repetitive elements. Four weeks after irradiation, these changes were primarily confined to HPSCs and were exhibited as dose-dependent hypermethylation of LINE1 and SINE B1 repetitive elements [4.2-fold increase in LINE1 (P < 0.001) and 7.6-fold increase in SINE B1 (P < 0.01) after exposure to 0.4 Gy; n = 5]. Epigenetic alterations were persistent and detectable for at least 22 weeks after exposure, when significant loss of global DNA hypomethylation (1.9-fold, P < 0.05), decreased expression of Dnmt1 (1.9-fold, P < 0.01), and increased expression of LINE1 and SINE B1 repetitive elements (2.8-fold, P < 0.001 for LINE1 and 1.9-fold, P < 0.05 for SINE B1; n = 5) were observed after exposure to 0.4 Gy. In contrast, exposure to (56)Fe ions did not result in accumulation of increased production of reactive oxygen species (ROS) and DNA damage, exhibited as DNA strand breaks. Furthermore, no significant alterations in cellular senescence and apoptosis were detected in HPSCs after exposure to (56)Fe-ion radiation. These findings suggest that epigenetic reprogramming is possibly involved in the development of radiation-induced genomic instability and thus, may have a causative role in the development of AML.
Collapse
Affiliation(s)
- Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Lijian Shao
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Jianhui Chang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Wei Feng
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Yingying Wang
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Antiño R Allen
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Department of Neurological Surgery, University of California San Francisco, San Francisco, California
| | - Jennifer Turner
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon
| | - Blair Stewart
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon
| | - Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon.,Department of Neurology, ONPRC, Oregon Health and Science University, Portland, Oregon.,Department of Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, Oregon
| | - Daohong Zhou
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|