294 results on '"William H. McBride"'
Search Results
2. Data from Radiotherapy Decreases Vascular Density and Causes Hypoxia with Macrophage Aggregation in TRAMP-C1 Prostate Tumors
- Author
-
Ji-Hong Hong, William H. McBride, Chung-Chi Lee, Shih-Ming Jung, Chien-Sheng Tsai, Chun-Chieh Wang, Chi-Shiun Chiang, and Fang-Hsin Chen
- Abstract
Purpose: To investigate how single or fractionated doses of radiation change the microenvironment in transgenic adenocarcinoma of the mouse prostate (TRAMP)-C1 tumors with respect to vascularity, hypoxia, and macrophage infiltrates.Experimental Design: Murine prostate TRAMP-C1 tumors were grown in C57BL/6J mice to 4 mm tumor diameter and were irradiated with either 25 Gy in a single dose or 60 Gy in 15 fractions. Changes in vascularity, hypoxia, and macrophage infiltrates were assessed by immunohistochemistry and molecular assays.Results: Tumor growth was delayed for 1 week after both radiation schedules. Tumor microvascular density (MVD) progressively decreased over a 3-week period to nadirs of 25% and 40% of unirradiated tumors for single or fractionated treatment, respectively. In accord with the decrease in MVDs, mRNA levels of endothelial markers, such as CD31, endoglin, and TIE, decreased over the same time period after irradiation. Central dilated vessels developed surrounded by avascularized hypoxic regions that became infiltrated with aggregates of CD68+ tumor-associated macrophages, reaching a maximum at 3 weeks after irradiation. Necrotic regions decreased and were more dispersed.Conclusion: Irradiation of TRAMP-C1 tumors with either single or fractionated doses decreases MVD, leading to the development of disperse chronic hypoxic regions, which are infiltrated with CD68+ tumor-associated macrophages. Approaches to interfere in the development of these effects are promising strategies to enhance the efficacy of cancer radiotherapy.
- Published
- 2023
- Full Text
- View/download PDF
3. Data from Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer
- Author
-
William H. McBride, Dörthe Schaue, Sandra Demaria, Begoña Comin-Anduix, John A. Glaspy, Sara Hurvitz, James W. Sayre, Kym F. Faull, Lin Hwang, Carol Felix, Josephine A. Ratikan, Mike W. Xie, Xiaochun Li, Judith D. Goldberg, Sylvia Adams, Percy Lee, and Silvia C. Formenti
- Abstract
Purpose: This study examined the feasibility, efficacy (abscopal effect), and immune effects of TGFβ blockade during radiotherapy in metastatic breast cancer patients.Experimental Design: Prospective randomized trial comparing two doses of TGFβ blocking antibody fresolimumab. Metastatic breast cancer patients with at least three distinct metastatic sites whose tumor had progressed after at least one line of therapy were randomized to receive 1 or 10 mg/kg of fresolimumab, every 3 weeks for five cycles, with focal radiotherapy to a metastatic site at week 1 (three doses of 7.5 Gy), that could be repeated to a second lesion at week 7. Research bloods were drawn at baseline, week 2, 5, and 15 to isolate PBMCs, plasma, and serum.Results: Twenty-three patients were randomized, median age 57 (range 35–77). Seven grade 3/4 adverse events occurred in 5 of 11 patients in the 1 mg/kg arm and in 2 of 12 patients in the 10 mg/kg arm, respectively. Response was limited to three stable disease. At a median follow up of 12 months, 20 of 23 patients are deceased. Patients receiving the 10 mg/kg had a significantly higher median overall survival than those receiving 1 mg/kg fresolimumab dose [hazard ratio: 2.73 with 95% confidence interval (CI), 1.02–7.30; P = 0.039]. The higher dose correlated with improved peripheral blood mononuclear cell counts and a striking boost in the CD8 central memory pool.Conclusions: TGFβ blockade during radiotherapy was feasible and well tolerated. Patients receiving the higher fresolimumab dose had a favorable systemic immune response and experienced longer median overall survival than the lower dose group. Clin Cancer Res; 24(11); 2493–504. ©2018 AACR.
- Published
- 2023
- Full Text
- View/download PDF
4. S1-4 from Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer
- Author
-
William H. McBride, Dörthe Schaue, Sandra Demaria, Begoña Comin-Anduix, John A. Glaspy, Sara Hurvitz, James W. Sayre, Kym F. Faull, Lin Hwang, Carol Felix, Josephine A. Ratikan, Mike W. Xie, Xiaochun Li, Judith D. Goldberg, Sylvia Adams, Percy Lee, and Silvia C. Formenti
- Abstract
Supplementary Table S1. Antibodies and dextramers used for flow cytometric analysis Supplementary Table S2. Patients with one or more treatment positively-related (maximum grade possible, probable, definite) AEs by treatment arm Supplementary Table S3. Levels of circulating tumor-specific CD8+ T cells with reactivity for JARID1B, Muc1 or Her2 in breast cancer patients with HLA-A*0201 status Supplementary Table S4. Humoral immune responses were not significantly affected following TGFβ blockade and radiation Supplementary Figure S1: Study schema Supplementary Figure S2. Gating strategy in panel 1. Supplementary Figure S3. Gating strategy in panel 2. Supplementary Figure S4. Changes in PBMC, survivin- and JARID1B-reactive CD8 T cells in response to TGFβ blockade and radiation. Supplementary Figure S5. Diametrically opposing dynamics in the myeloid suppressor compartment versus the suppressor T cell pool in response to TGFβ blockade and radiation. Supplementary Figure S6. Changes in plasma levels of tryptophan and kynurenine in response to TGFβ blockade and radiation.
- Published
- 2023
- Full Text
- View/download PDF
5. Supplementary Figure S1 from T-Cell Responses to Survivin in Cancer Patients Undergoing Radiation Therapy
- Author
-
William H. McBride, Karin Haustermans, Annelies Debucquoy, James W. Sayre, Lee Goodglick, Li Zhang, Antoni Ribas, Begonya Comin-Anduix, and Dörthe Schaue
- Abstract
Supplementary Figure S1 from T-Cell Responses to Survivin in Cancer Patients Undergoing Radiation Therapy
- Published
- 2023
- Full Text
- View/download PDF
6. Data from T-Cell Responses to Survivin in Cancer Patients Undergoing Radiation Therapy
- Author
-
William H. McBride, Karin Haustermans, Annelies Debucquoy, James W. Sayre, Lee Goodglick, Li Zhang, Antoni Ribas, Begonya Comin-Anduix, and Dörthe Schaue
- Abstract
Purpose: The goal of this study was to determine if radiation therapy (RT) of human cancer enhances or diminishes tumor-specific T-cell reactivity. This is important if immunotherapy is to be harnessed to improve the outcome of cancer radiotherapy.Experimental Design: Lymphocytes were isolated from colorectal cancer (CRC) patients before, during, and after presurgical chemoradiotherapy. Similar samples were taken from prostate cancer patients receiving standard RT. The level of CD8+ T cells capable of binding tetramers for the tumor-associated antigen survivin, which is overexpressed in both cancer types, was enumerated in HLA-A*0201 patient samples. CD4+, CD25high, Foxp3+ cells were also enumerated to evaluate therapy-induced changes in Tregulatory cells. For CRC patients, most of whom were enrolled in a clinical trial, pathologic response data were available, as well as biopsy and resection specimens, which were stained for cytoplasmic and intranuclear survivin.Results: Survivin-specific CD8+ T lymphocytes were detected in the peripheral blood of CRC and prostate cancer patients and increased after therapy in some, but not all, patients. Increases were more common in CRC patients whose tumor was downstaged after chemoradiotherapy. Biopsy specimens from this cohort generally had higher nuclear to cytoplasmic survivin expression. Tregulatory cells generally increased in the circulation following therapy but only in CRC patients.Conclusion: This study indicates that RT may increase the likelihood of some cancer patients responding to immunotherapy and lays a basis for future investigations aimed at combining radiation and immunotherapy.
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Figures S1-S3 from Radiotherapy Decreases Vascular Density and Causes Hypoxia with Macrophage Aggregation in TRAMP-C1 Prostate Tumors
- Author
-
Ji-Hong Hong, William H. McBride, Chung-Chi Lee, Shih-Ming Jung, Chien-Sheng Tsai, Chun-Chieh Wang, Chi-Shiun Chiang, and Fang-Hsin Chen
- Abstract
Supplementary Figures S1-S3 from Radiotherapy Decreases Vascular Density and Causes Hypoxia with Macrophage Aggregation in TRAMP-C1 Prostate Tumors
- Published
- 2023
- Full Text
- View/download PDF
8. Supplementary Figure 2 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
Supplementary Figure 2 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Figure Legends 1-4 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
Supplementary Figure Legends 1-4 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
The transcription factor NF-E2-related factor 2 (Nrf2) binds the antioxidant DNA response element (ARE) to activate important cellular cytoprotective defense systems. Recently several types of cancers have been shown to overexpress Nrf2, but its role in the cellular response to radiation therapy has yet to be fully determined. In this study, we report that single doses of ionizing radiation from 2 to 8 Gy activate ARE-dependent transcription in breast cancer cells in a dose-dependent manner, but only after a delay of five days. Clinically relevant daily dose fractions of radiation also increased ARE-dependent transcription, but again only after five days. Downstream activation of Nrf2-ARE-dependent gene and protein markers, such as heme oxygenase-1, occurred, whereas Nrf2-deficient fibroblasts were incapable of these responses. Compared with wild-type fibroblasts, Nrf2-deficient fibroblasts had relatively high basal levels of reactive oxygen species that increased greatly five days after radiation exposure. Further, in vitro clonogenic survival assays and in vivo sublethal whole body irradiation tests showed that Nrf2 deletion increased radiation sensitivity, whereas Nrf2-inducing drugs did not increase radioresistance. Our results indicate that the Nrf2-ARE pathway is important to maintain resistance to irradiation, but that it operates as a second-tier antioxidant adaptive response system activated by radiation only under specific circumstances, including those that may be highly relevant to tumor response during standard clinical dose-fractionated radiation therapy. Cancer Res; 70(21); 8886–95. ©2010 AACR.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figure 4 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
Supplementary Figure 4 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure 1 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
Supplementary Figure 1 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Published
- 2023
- Full Text
- View/download PDF
13. Supplementary Figure 3 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Author
-
William H. McBride, Lynn Hlatky, Heather Szelag, Josephine Ratikan, Lorenza Della Donna, Chann Lagadec, Tiffany M. Phillips, Erina Vlashi, Andrew J. Norris, Kwanghee Kim, and J. Tyson McDonald
- Abstract
Supplementary Figure 3 from Ionizing Radiation Activates the Nrf2 Antioxidant Response
- Published
- 2023
- Full Text
- View/download PDF
14. NRF2 Mediates Cellular Resistance to Transformation, Radiation, and Inflammation in Mice
- Author
-
Dörthe Schaue, Ewa D. Micewicz, Josephine A. Ratikan, Keisuke S. Iwamoto, Erina Vlashi, J. Tyson McDonald, and William H. McBride
- Subjects
Physiology ,Clinical Biochemistry ,immune polarization ,Cell Biology ,ionizing radiation ,Molecular Biology ,Biochemistry ,Nrf2 ,NF-κB - Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is recognized as a master transcription factor that regulates expression of numerous detoxifying and antioxidant cytoprotective genes. In fact, models of NRF2 deficiency indicate roles not only in redox regulation, but also in metabolism, inflammatory/autoimmune disease, cancer, and radioresistancy. Since ionizing radiation (IR) generates reactive oxygen species (ROS), it is not surprising it activates NRF2 pathways. However, unexpectedly, activation is often delayed for many days after the initial ROS burst. Here, we demonstrate that, as assayed by γ-H2AX staining, rapid DNA double strand break (DSB) formation by IR in primary mouse Nrf2–/– MEFs was not affected by loss of NRF2, and neither was DSB repair to any great extent. In spite of this, basal and IR-induced transformation was greatly enhanced, suggesting that NRF2 protects against late IR-induced genomic instability, at least in murine MEFs. Another possible IR- and NRF2-related event that could be altered is inflammation and NRF2 deficiency increased IR-induced NF-κB pro-inflammatory responses mostly late after exposure. The proclivity of NRF2 to restrain inflammation is also reflected in the reprogramming of tumor antigen-specific lymphocyte responses in mice where Nrf2 k.o. switches Th2 responses to Th1 polarity. Delayed NRF2 responses to IR may be critical for the immune transition from prooxidant inflammation to antioxidant healing as well as in driving cellular radioresistance and survival. Targeting NRF2 to reprogram immunity could be of considerable therapeutic benefit in radiation and immunotherapy.
- Published
- 2022
- Full Text
- View/download PDF
15. Abstract 2325: IRF1 displays opposing tumor cell-intrinsic and -extrinsic roles in anti-tumor immunity
- Author
-
Prabhat Kumar Purbey, Manash K. Paul, Keisuke S. Iwamoto, Allison Daly, Joowon Seo, Ameya S. Champhekar, Katie Campbell, Dorthe Schaue, William H. McBride, Steven M. Dubinett, Antoni Ribas, Stephen T. Smale, and Philip O. Scumpia
- Subjects
Cancer Research ,Oncology - Abstract
Interferons (IFNs), including IFN-α/β and IFN-γ, are multifaceted cytokines critical for antitumor immunity but contribute to T cell exhaustion through immune checkpoints. Recently, tumor cell expression of IRF1 was shown to enhance antitumor immunity, but a comprehensive evaluation of how IRF1 regulates IFN responses in the tumor and host has not been performed. Here, we show that Irf1-/- mice display enhanced tumor growth of WT and Irf1-/- tumors, as they fail to recruit sufficient NK cells and cytotoxic T cell (CTL). However, Irf1-/- tumors in wild-type mice displayed rapid immunogenic control, expansion of intratumoral lymphoid cells with enhanced effector programs and diminished exhaustion programs when compared to WT tumors. Mechanistically, IRF1 positively regulates the expression of distinct subsets of anti-tumor immunity suppression genes including the CD274/PD-L1, TRAIL, and IDO-1 checkpoints but not the IFN-inducible chemokines CXCL9-11. Surprisingly, control of Irf1-/- tumors required host IFN-α/β signaling but not IFN-γ signaling, while PD-L1 overexpression only weakly restored tumor growth. Thus, IRF1 selectively regulates the effects of IFNs on tumor cells and the tumor microenvironment and may be targeted to boost anti-tumor immunity. Citation Format: Prabhat Kumar Purbey, Manash K. Paul, Keisuke S. Iwamoto, Allison Daly, Joowon Seo, Ameya S. Champhekar, Katie Campbell, Dorthe Schaue, William H. McBride, Steven M. Dubinett, Antoni Ribas, Stephen T. Smale, Philip O. Scumpia. IRF1 displays opposing tumor cell-intrinsic and -extrinsic roles in anti-tumor immunity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 2325.
- Published
- 2023
- Full Text
- View/download PDF
16. Radiation‐induced tissue damage and response
- Author
-
Dörthe Schaue and William H. McBride
- Subjects
0301 basic medicine ,clonogen ,Radiobiology ,Time Factors ,Context (language use) ,Radiation Tolerance ,Pathology and Forensic Medicine ,Ionizing radiation ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,Medicine ,oxidative stress ,acute and late responsessenescenceautophagyinflammationregeneration ,Humans ,Invited Reviews ,Radiosensitivity ,Clonogenic assay ,Invited Review ,business.industry ,Regeneration (biology) ,Bone Marrow Stem Cell ,Dose-Response Relationship, Radiation ,stem cell ,030104 developmental biology ,Acute Radiation Syndrome ,030220 oncology & carcinogenesis ,Cancer research ,Stem cell ,business ,ionizing radiation ,DNA Damage - Abstract
Normal tissue responses to ionizing radiation have been a major subject for study since the discovery of X‐rays at the end of the 19th century. Shortly thereafter, time–dose relationships were established for some normal tissue endpoints that led to investigations into how the size of dose per fraction and the quality of radiation affected outcome. The assessment of the radiosensitivity of bone marrow stem cells using colony‐forming assays by Till and McCulloch prompted the establishment of in situ clonogenic assays for other tissues that added to the radiobiology toolbox. These clonogenic and functional endpoints enabled mathematical modeling to be performed that elucidated how tissue structure, and in particular turnover time, impacted clinically relevant fractionated radiation schedules. More recently, lineage tracing technology, advanced imaging and single cell sequencing have shed further light on the behavior of cells within stem, and other, cellular compartments, both in homeostasis and after radiation damage. The discovery of heterogeneity within the stem cell compartment and plasticity in response to injury have added new dimensions to the consideration of radiation‐induced tissue damage. Clinically, radiobiology of the 20th century garnered wisdom relevant to photon treatments delivered to a fairly wide field at around 2 Gy per fraction, 5 days per week, for 5–7 weeks. Recently, the scope of radiobiology has been extended by advances in technology, imaging and computing, as well as by the use of charged particles. These allow radiation to be delivered more precisely to tumors while minimizing the amount of normal tissue receiving high doses. One result has been an increase in the use of schedules with higher doses per fraction given in a shorter time frame (hypofractionation). We are unable to cover these new technologies in detail in this review, just as we must omit low‐dose stochastic effects, and many aspects of dose, dose rate and radiation quality. We argue that structural diversity and plasticity within tissue compartments provides a general context for discussion of most radiation responses, while acknowledging many omissions. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
- Published
- 2020
17. Immune Networks in the Context of Low Dose Ionizing Radiation
- Author
-
Dörthe Schaue, Keisuke S. Iwamoto, and William H. McBride
- Published
- 2022
- Full Text
- View/download PDF
18. BCN057, a Modulator of GSK3β, Induces KRAS G12D Mutant Pancreatic Cancer Cell Death
- Author
-
Adrian M. Gomez, Payel Bhanja, Andrew J. Norris, Elizabeth M. Singer, Subhrajit Saha, William H. McBride, Rishi Mann Chugh, Julian P. Whitelegge, and Lucy W Gao
- Subjects
business.industry ,LGR5 ,Synthetic lethality ,medicine.disease ,medicine.disease_cause ,Gastrointestinal epithelium ,Intestinal epithelium ,medicine.anatomical_structure ,Pancreatic cancer ,medicine ,Cancer research ,KRAS ,Stem cell ,business ,Pancreas - Abstract
Effective treatment for Pancreatic Cancer remains a major challenge due to its resistance to radiation/chemotherapy and poor drug permeability. Moreover, treatment induced normal tissue toxicity, mainly to the duodenum and gastrointestinal epithelium, is common and is a dose limiting event, while toxicity to the pancreas is relatively rare1–3. Gastrointestinal toxicity, however, often results in interruption, reduction or premature withdrawal of anti-cancer therapy which is a very significant factor impacting the overall survival of patients being treated. Therefore, development of a therapeutic strategy to selectively sensitize tumor tissue without inducing normal tissue toxicity is important. In this manuscript, we show that the novel small molecule BCN057 can modulate chemo-sensitivity of oncogenic RAS pancreatic cancer cells while conversely protecting normal intestinal epithelium from off target toxicity. In particular, BCN 057 protects Lgr5 positive intestinal stem cells, thereby preserving barrier function. Further, it is demonstrated that BCN057 inhibits GSK3β and thereby induces a pro-apoptotic phosphorylation pattern on c-Jun in KRAS G12D mutant pancreatic cancer cells (Panc-1) leading to the restoration of PTEN expression and consequent apoptosis. This appears to be a new mechanistic observation for the oncogenic RAS phenotype. Lastly, concurrent with its GSK3β inhibition, BCN057 is a small molecule inhibitor of PD-1 expression on human T-lymphocytes co-cultured with human pancreatic cancer cells. In summary, BCN057 can promote synthetic lethality specifically to malignant cells and therefore should be considered to improve the therapeutic ratio in pancreatic and epithelial cancer treatment in conjunction with chemotherapy and radiation.
- Published
- 2021
- Full Text
- View/download PDF
19. Immunotherapy in Combination With Radiation Therapy
- Author
-
Jean Philippe Nesseler, William H. McBride, and Dörthe Schaue
- Published
- 2021
- Full Text
- View/download PDF
20. PTPσ inhibitors promote hematopoietic stem cell regeneration
- Author
-
Michelle Li, Liman Zhao, Heather A. Himburg, Tiancheng Fang, Yurun Zhang, Katherine Pohl, Martina Roos, Christina M. Termini, William H. McBride, Mamle Quarmyne, Robert Damoiseaux, Hyo Jin Gim, Michael E. Jung, Xiao Yan, John P. Chute, Julian P. Whitelegge, Emelyne Diers, and Jenny Kan
- Subjects
0301 basic medicine ,rac1 GTP-Binding Protein ,rho GTP-Binding Proteins ,Antimetabolites ,General Physics and Astronomy ,Receptor-Like Protein Tyrosine Phosphatases ,Apoptosis ,02 engineering and technology ,Stem cells ,Regenerative Medicine ,Mice ,Stem Cell Research - Nonembryonic - Human ,Tyrosine ,Enzyme Inhibitors ,lcsh:Science ,Cancer ,Multidisciplinary ,Radiation ,Chemistry ,Haematopoietic stem cells ,Receptor-Like Protein Tyrosine Phosphatases, Class 2 ,Hematopoietic stem cell ,Hematology ,021001 nanoscience & nanotechnology ,Antineoplastic ,3. Good health ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,5.1 Pharmaceuticals ,Systemic administration ,Stem Cell Research - Nonembryonic - Non-Human ,Fluorouracil ,Development of treatments and therapeutic interventions ,0210 nano-technology ,Antimetabolites, Antineoplastic ,1.1 Normal biological development and functioning ,Science ,Allosteric regulation ,bcl-X Protein ,RAC1 ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Allosteric Regulation ,Underpinning research ,medicine ,Animals ,Humans ,Regeneration ,Transplantation ,Regeneration (biology) ,General Chemistry ,Class 2 ,Stem Cell Research ,Hematopoietic Stem Cells ,030104 developmental biology ,lcsh:Q - Abstract
Receptor type protein tyrosine phosphatase-sigma (PTPσ) is primarily expressed by adult neurons and regulates neural regeneration. We recently discovered that PTPσ is also expressed by hematopoietic stem cells (HSCs). Here, we describe small molecule inhibitors of PTPσ that promote HSC regeneration in vivo. Systemic administration of the PTPσ inhibitor, DJ001, or its analog, to irradiated mice promotes HSC regeneration, accelerates hematologic recovery, and improves survival. Similarly, DJ001 administration accelerates hematologic recovery in mice treated with 5-fluorouracil chemotherapy. DJ001 displays high specificity for PTPσ and antagonizes PTPσ via unique non-competitive, allosteric binding. Mechanistically, DJ001 suppresses radiation-induced HSC apoptosis via activation of the RhoGTPase, RAC1, and induction of BCL-XL. Furthermore, treatment of irradiated human HSCs with DJ001 promotes the regeneration of human HSCs capable of multilineage in vivo repopulation. These studies demonstrate the therapeutic potential of selective, small-molecule PTPσ inhibitors for human hematopoietic regeneration., Protein tyrosine phosphatase sigma (PTPσ) deficient haematopoietic stem cells (HSCs) demonstrate increased engraftment following transplantation. Here the authors identify a small molecule inhibitor of PTPσ that promotes murine and human haematopoietic stem cell regeneration via induction of the RAC pathway and BCL-XL.
- Published
- 2019
21. Baseline T cell dysfunction by single cell network profiling in metastatic breast cancer patients
- Author
-
Dörthe Schaue, William H. McBride, Erik Evensen, Percy Lee, Judith D. Goldberg, Xiaochun Li, Neha Dixit, Claire Vanpouille-Box, Silvia C. Formenti, Rachael E. Hawtin, and Sandra Demaria
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,B7-H1 Antigen ,0302 clinical medicine ,Programmed Death-1 (PD-1) ,Transforming Growth Factor beta ,Receptors ,Monoclonal ,Immunology and Allergy ,Cytotoxic T cell ,Programmed Death-1 ,T cell receptor (TCR) signaling ,Humanized ,Cancer ,biology ,CD28 ,Middle Aged ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Combined Modality Therapy ,Metastatic breast cancer ,3. Good health ,medicine.anatomical_structure ,Antigen ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,Immunotherapy ,Adult ,medicine.medical_specialty ,T cell ,CD3 ,Clinical Trials and Supportive Activities ,Immunology ,Receptors, Antigen, T-Cell ,Short Report ,Breast Neoplasms ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,lcsh:RC254-282 ,Antibodies ,Young Adult ,03 medical and health sciences ,Clinical Research ,Internal medicine ,Breast Cancer ,medicine ,Humans ,Aged ,Pharmacology ,Radiotherapy ,business.industry ,T-Cell ,medicine.disease ,Brain Disorders ,Blockade ,030104 developmental biology ,biology.protein ,business ,CD8 - Abstract
Background We previously reported the results of a multicentric prospective randomized trial of chemo-refractory metastatic breast cancer patients testing the efficacy of two doses of TGFβ blockade during radiotherapy. Despite a lack of objective responses to the combination, patients who received a higher dose of TGFβ blocking antibody fresolimumab had a better overall survival when compared to those assigned to lower dose (hazard ratio of 2.73, p = 0.039). They also demonstrated an improved peripheral blood mononuclear cell (PBMC) counts and increase in the CD8 central memory pool. We performed additional analysis on residual PBMC using single cell network profiling (SCNP). Methods The original trial randomized metastatic breast cancer patients to either 1 or 10 mg/kg of fresolimumab, every 3 weeks for 5 cycles, combined with radiotherapy to a metastatic site at week 1 and 7 (22.5 Gy given in 3 doses of 7.5 Gy). Trial immune monitoring results were previously reported. In 15 patients with available residual blood samples, additional functional studies were performed, and compared with data obtained in parallel from seven healthy female donors (HD): SCNP was applied to analyze T cell receptor (TCR) modulated signaling via CD3 and CD28 crosslinking and measurement of evoked phosphorylation of AKT and ERK in CD4 and CD8 T cell subsets defined by PD-1 expression. Results At baseline, a significantly higher level of expression (p
- Published
- 2019
- Full Text
- View/download PDF
22. Irradiation to Improve the Response to Immunotherapeutic Agents in Glioblastomas
- Author
-
Tania Kaprealian, Simone P. Niclou, Philippe Nickers, Mi-Heon Lee, Dörthe Schaue, William H. McBride, and J.P. Nesseler
- Subjects
Central Nervous System ,lcsh:Medical physics. Medical radiology. Nuclear medicine ,medicine.medical_treatment ,lcsh:R895-920 ,lcsh:RC254-282 ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Therapeutic approach ,0302 clinical medicine ,Immune system ,medicine ,Radiology, Nuclear Medicine and imaging ,Temozolomide ,biology ,business.industry ,Immunotherapy ,Transforming growth factor beta ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immune checkpoint ,3. Good health ,Clinical trial ,Radiation therapy ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business ,medicine.drug - Abstract
Purpose Glioblastoma (GBM) remains an incurable disease despite extensive treatment with surgical resection, irradiation, and temozolomide. In line with many other forms of aggressive cancers, GBM is currently under consideration as a target for immunotherapy. However, GBM tends to be nonimmunogenic and exhibits a microenvironment with few or no effector T cells, a relatively low nonsynonymous somatic mutational load, and a low predicted neoantigen burden. GBM also exploits a multitude of immunosuppressive strategies. Methods and Materials A number of immunotherapeutic approaches have been tested with disappointing results. A rationale exists to combine immunotherapy and radiation therapy, which can induce an immunogenic form of cell death with T-cell activation and tumor infiltration. Results Various immunotherapy agents, including immune checkpoint modulators, transforming growth factor beta receptor inhibitors, and indoleamine-2,3-dioxygenase inhibitors, have been evaluated with irradiation in preclinical GBM models, with promising results, and are being further tested in clinical trials. Conclusions This review aims to present the basic rationale behind this emerging complementary therapeutic approach in GBM, appraise the current preclinical and clinical data, and discuss the future challenges in improving the antitumor immune response.
- Published
- 2019
23. Are animal models a necessity for acute radiation syndrome drug discovery?
- Author
-
Dörthe Schaue and William H. McBride
- Subjects
Acute Radiation Syndrome ,business.industry ,Drug discovery ,Models, Animal ,Drug Discovery ,Animals ,Humans ,Medicine ,Radiation-Protective Agents ,Bioinformatics ,business ,Article - Published
- 2019
- Full Text
- View/download PDF
24. Classes of Drugs that Mitigate Radiation Syndromes
- Author
-
Ewa D. Micewicz, Robert D. Damoiseaux, Gang Deng, Adrian Gomez, Keisuke S. Iwamoto, Michael E. Jung, Christine Nguyen, Andrew J. Norris, Josephine A. Ratikan, Piotr Ruchala, James W. Sayre, Dörthe Schaue, Julian P. Whitelegge, and William H. McBride
- Subjects
Drug ,High-throughput screening ,media_common.quotation_subject ,Drug availability ,radiation mitigators ,RM1-950 ,Pharmacology ,Ionizing radiation ,Vaccine Related ,gastro-intestinal acute radiation syndrome ,Rare Diseases ,In vivo ,Biodefense ,High throughput screening ,Pharmacology (medical) ,Radiation syndromes ,Cancer ,Original Research ,media_common ,Chemistry ,Prevention ,Pharmacology and Pharmaceutical Sciences ,Small molecule ,Bioavailability ,Orphan Drug ,delayed effects of radiation exposure ,hematopoietic acute radiation syndrome ,5.1 Pharmaceuticals ,Therapeutics. Pharmacology ,Development of treatments and therapeutic interventions ,Biotechnology ,multi-organ disease syndrome - Abstract
We previously reported several vignettes on types and classes of drugs able to mitigate acute and, in at least one case, late radiation syndromes in mice. Most of these had emerged from high throughput screening (HTS) of bioactive and chemical drug libraries using ionizing radiation-induced lymphocytic apoptosis as a readout. Here we report the full analysis of the HTS screen of libraries with 85,000 small molecule chemicals that identified 220 “hits.” Most of these hits could be allocated by maximal common substructure analysis to one of 11 clusters each containing at least three active compounds. Further screening validated 23 compounds as being most active; 15 of these were cherry-picked based on drug availability and tested for their ability to mitigate acute hematopoietic radiation syndrome (H-ARS) in mice. Of these, five bore a 4-nitrophenylsulfonamide motif while 4 had a quinoline scaffold. All but two of the 15 significantly (p < 0.05) mitigated H-ARS in mice. We had previously reported that the lead 4-(nitrophenylsulfonyl)-4-phenylpiperazine compound (NPSP512), was active in mitigating multiple acute and late radiation syndromes in mice of more than one sex and strain. Unfortunately, the formulation of this drug had to be changed for regulatory reasons and we report here on the synthesis and testing of active analogs of NPSP512 (QS1 and 52A1) that have increased solubility in water and in vivo bioavailability while retaining mitigator activity against H-ARS (p < 0.0001) and other radiation syndromes. The lead quinoline 057 was also active in multiple murine models of radiation damage. Taken together, HTS of a total of 150,000 bioactive or chemical substances, combined with maximal common substructure analysis has resulted in the discovery of diverse groups of compounds that can mitigate H-ARS and at least some of which can mitigate multiple radiation syndromes when given starting 24 h after exposure. We discuss what is known about how these agents might work, and the importance of formulation and bioavailability.
- Published
- 2021
- Full Text
- View/download PDF
25. Weak Magnetic Fields Enhance the Efficacy of Radiation Therapy
- Author
-
Robert E. Sandstrom, S. Robin Elgart, Michael L. Steinberg, Daniel A. Low, Ke Sheng, William H. McBride, Mark Bryan, Yue Liu, and Keisuke S. Iwamoto
- Subjects
medicine.medical_treatment ,Radical ,R895-920 ,Context (language use) ,Radiation ,030218 nuclear medicine & medical imaging ,Ionizing radiation ,03 medical and health sciences ,Medical physics. Medical radiology. Nuclear medicine ,0302 clinical medicine ,In vivo ,medicine ,Scientific Article ,Radiology, Nuclear Medicine and imaging ,RC254-282 ,Cancer ,chemistry.chemical_classification ,Reactive oxygen species ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Radiation therapy ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Biophysics ,business - Abstract
Purpose The clinical efficacy of radiation therapy is mechanistically linked to ionization-induced free radicals that cause cell and tissue injury through direct and indirect mechanisms. Free radical reaction dynamics are influenced by many factors and can be manipulated by static weak magnetic fields (WMF) that perturb singlet-triplet state interconversion. Our study exploits this phenomenon to directly increase ionizing radiation (IR) dose absorption in tumors by combining WMF with radiotherapy as a new and effective method to improve treatment. Methods and Materials Coils were custom made to produce both homogeneous and gradient magnetic fields. The gradient coil enabled simultaneous in vitro assessment of free radical/reactive oxygen species (ROS) reactivity across multiple field strengths from 6 to 66 G. First, increases in ionizing radiation-induced free radical concentrations using oxidant-sensitive fluorescent dyes in a cell-free system were measured and verified. Next, human and murine cancer cell lines were evaluated in in vitro and in vivo models following exposure to clinically relevant doses of ionizing radiation in combination with WMF. Results Cellular responses to IR and WMF were field strength- and cell line-dependent. WMF was able to enhance IR effects on ROS formation, DNA double strand break formation, cell death, and tumor growth. Conclusions We demonstrate that the external presence of a magnetic field enhances radiation-induced cancer cell injury and death in vitro and in vivo. The effect extends beyond the timeframe when free radicals are induced in the presence of radiation into the window when endogenous free radicals are produced and therefore extends the applicability of this novel adjunct to cancer therapy in the context of radiation treatment.
- Published
- 2021
26. Low-Dose Radiation Therapy (LDRT) for COVID-19: Benefits or Risks?
- Author
-
Bhadrasain Vikram, Dale J. Hu, Mansoor M. Ahmed, Ralph R. Weichselbaum, David G. Kirsch, Andrea L. DiCarlo, Silvia C. Formenti, Gayle E. Woloschak, Kathryn D. Held, Chandan Guha, C. Norman Coleman, Arnab Chakravarti, Dörthe Schaue, William H. McBride, Jeffrey C. Buchsbaum, Sunil Krishnan, Pataje G. S. Prasanna, Brian Marples, Wolfgang W. Leitner, Francis A. Cucinotta, Mohammad K. Khan, Shahin Rafii, Julie M. Sullivan, Elad Sharon, and Minesh P. Mehta
- Subjects
Risk ,medicine.medical_specialty ,Radiobiology ,Coronavirus disease 2019 (COVID-19) ,medicine.medical_treatment ,Pneumonia, Viral ,Biophysics ,MEDLINE ,Radiation Dosage ,Article ,030218 nuclear medicine & medical imaging ,law.invention ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Randomized controlled trial ,law ,Radiation oncology ,medicine ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Medical physics ,Pandemics ,Clinical Trials as Topic ,Radiation ,business.industry ,COVID-19 ,Radiotherapy Dosage ,Radiation therapy ,030220 oncology & carcinogenesis ,Low Dose Radiation Therapy ,Radiation protection ,business ,Coronavirus Infections - Abstract
The limited impact of treatments for COVID-19 has stimulated several phase 1 clinical trials of whole-lung low-dose radiation therapy (LDRT; 0.3-1.5 Gy) that are now progressing to phase 2 randomized trials worldwide. This novel but unconventional use of radiation to treat COVID-19 prompted the National Cancer Institute, National Council on Radiation Protection and Measurements and National Institute of Allergy and Infectious Diseases to convene a workshop involving a diverse group of experts in radiation oncology, radiobiology, virology, immunology, radiation protection and public health policy. The workshop was held to discuss the mechanistic underpinnings, rationale, and preclinical and emerging clinical studies, and to develop a general framework for use in clinical studies. Without refuting or endorsing LDRT as a treatment for COVID-19, the purpose of the workshop and this review is to provide guidance to clinicians and researchers who plan to conduct preclinical and clinical studies, given the limited available evidence on its safety and efficacy.
- Published
- 2020
27. Three discipline collaborative radiation therapy (3DCRT) special debate: We should treat all cancer patients with hypofractionation
- Author
-
Emilie Soisson, Michael M. Dominello, Jay Burmeister, Samantha J. Van Nest, Kathryn E. Huber, William H. McBride, Y Liao, Michael C. Joiner, and Michael D. Green
- Subjects
medicine.medical_specialty ,Radiation ,business.industry ,medicine.medical_treatment ,MEDLINE ,Cancer ,medicine.disease ,Radiation therapy ,Neoplasms ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Medical physics ,Radiation Dose Hypofractionation ,Radiotherapy, Intensity-Modulated ,Radiotherapy, Conformal ,business ,Parallel Opposed Editorial ,Instrumentation - Published
- 2020
28. Flying by the seat of our pants: is low dose radiation therapy for COVID-19 an option?
- Author
-
Dörthe Schaue and William H. McBride
- Subjects
2019-20 coronavirus outbreak ,Reactive oxygen species metabolism ,Coronavirus disease 2019 (COVID-19) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Bioinformatics ,Article ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,Betacoronavirus ,0302 clinical medicine ,Pandemic ,Medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Pandemics ,Radiological and Ultrasound Technology ,biology ,business.industry ,SARS-CoV-2 ,COVID-19 ,Radiotherapy Dosage ,biology.organism_classification ,Radiology Nuclear Medicine and imaging ,030220 oncology & carcinogenesis ,Low Dose Radiation Therapy ,business ,Coronavirus Infections ,Reactive Oxygen Species - Abstract
The discovery of X-rays in 1895 had a dramatic impact on many aspects of society. One was the hope that it would be possible to treat some of the numerous infections and inflammatory conditions for...
- Published
- 2020
29. Identification of miRNA signatures associated with radiation-induced late lung injury in mice
- Author
-
Jason T. Lee, Ewa D. Micewicz, Agnes I. Lukaszewicz, Mark A. Starbird, Thomas A. Miller, Dörthe Schaue, William H. McBride, Jason Yamada-Hanff, Naresh Menon, Christine Nguyen, Keisuke S. Iwamoto, Josephine A. Ratikan, Claude J. Rogers, Tove Olafsen, and Amendola, Roberto
- Subjects
0301 basic medicine ,Male ,Pathology ,Pulmonology ,Physiology ,Cardiovascular ,Inbred C57BL ,Biochemistry ,Mice ,0302 clinical medicine ,Fibrosis ,Blood plasma ,Medicine and Health Sciences ,2.1 Biological and endogenous factors ,Tissue Distribution ,Aetiology ,Lung ,Mice, Inbred C3H ,Multidisciplinary ,Heart ,Animal Models ,Thorax ,Inbred C3H ,Body Fluids ,Nucleic acids ,Radiation Injuries, Experimental ,Infectious Diseases ,Heart Disease ,medicine.anatomical_structure ,Blood ,Experimental Organism Systems ,030220 oncology & carcinogenesis ,Medicine ,Female ,Anatomy ,Biotechnology ,Research Article ,medicine.medical_specialty ,General Science & Technology ,Science ,Mouse Models ,Lung injury ,Research and Analysis Methods ,Late Radiation Injury ,Blood Plasma ,03 medical and health sciences ,Experimental ,Rare Diseases ,Model Organisms ,Species Specificity ,White blood cell ,medicine ,Genetics ,Animals ,Humans ,Circulating MicroRNA ,Radiation Injuries ,Non-coding RNA ,Pneumonitis ,Proportional Hazards Models ,Natural antisense transcripts ,Biology and life sciences ,business.industry ,Prevention ,Myocardium ,medicine.disease ,Gene regulation ,Mice, Inbred C57BL ,Radiation Pneumonitis ,MicroRNAs ,Good Health and Well Being ,030104 developmental biology ,Cardiovascular Anatomy ,Animal Studies ,RNA ,Gene expression ,business ,Biomarkers ,Developmental Biology - Abstract
Acute radiation exposure of the thorax can lead to late serious, and even life-threatening, pulmonary and cardiac damage. Sporadic in nature, late complications tend to be difficult to predict, which prompted this investigation into identifying non-invasive, tissue-specific biomarkers for the early detection of late radiation injury. Levels of circulating microRNA (miRNA) were measured in C3H and C57Bl/6 mice after whole thorax irradiation at doses yielding approximately 70% mortality in 120 or 180 days, respectively (LD70/120 or 180). Within the first two weeks after exposure, weight gain slowed compared to sham treated mice along with a temporary drop in white blood cell counts. 52% of C3H (33 of 64) and 72% of C57Bl/6 (46 of 64) irradiated mice died due to late radiation injury. Lung and heart damage, as assessed by computed tomography (CT) and histology at 150 (C3H mice) and 180 (C57Bl/6 mice) days, correlated well with the appearance of a local, miRNA signature in the lung and heart tissue of irradiated animals, consistent with inherent differences in the C3H and C57Bl/6 strains in their propensity for developing radiation-induced pneumonitis or fibrosis, respectively. Radiation-induced changes in the circulating miRNA profile were most prominent within the first 30 days after exposure and included miRNA known to regulate inflammation and fibrosis. Importantly, early changes in plasma miRNA expression predicted survival with reasonable accuracy (88-92%). The miRNA signature that predicted survival in C3H mice, including miR-34a-5p, -100-5p, and -150-5p, were associated with pro-inflammatory NF-κB-mediated signaling pathways, whereas the signature identified in C57Bl/6 mice (miR-34b-3p, -96-5p, and -802-5p) was associated with TGF-β/SMAD signaling. This study supports the hypothesis that plasma miRNA profiles could be used to identify individuals at high risk of organ-specific late radiation damage, with applications for radiation oncology clinical practice or in the context of a radiological incident.
- Published
- 2020
30. Tumor Size Matters—Understanding Concomitant Tumor Immunity in the Context of Hypofractionated Radiotherapy with Immunotherapy
- Author
-
Christine Nguyen, Anusha Kalbasi, Tahmineh Romero, James Sayre, J.P. Nesseler, Mi-Heon Lee, Dörthe Schaue, William H. McBride, and Philippe Nickers
- Subjects
0301 basic medicine ,Cancer Research ,Myeloid ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Context (language use) ,Spleen ,lcsh:RC254-282 ,radiation therapy ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Cancer ,business.industry ,tumor size ,Immunotherapy ,programmed cell death protein 1 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Primary tumor ,Radiation therapy ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Immunization ,fibrosarcoma ,business ,CD8 - Abstract
The purpose of this study was to determine the dynamic contributions of different immune cell subsets to primary and abscopal tumor regression after hypofractionated radiation therapy (hRT) and the impact of anti-PD-1 therapy. A bilateral syngeneic FSA1 fibrosarcoma model was used in immunocompetent C3H mice, with delayed inoculation to mimic primary and microscopic disease. The effect of tumor burden on intratumoral and splenic immune cell content was delineated as a prelude to hRT on macroscopic T1 tumors with 3 fractions of 8 Gy while microscopic T2 tumors were left untreated. This was performed with and without systemic anti-PD-1. Immune profiles within T1 and T2 tumors and in spleen changed drastically with tumor burden in untreated mice with infiltrating CD4+ content declining, while the proportion of CD4+ Tregs rose. Myeloid cell representation escalated in larger tumors, resulting in major decreases in the lymphoid:myeloid ratios. In general, activation of Tregs and myeloid-derived suppressor cells allow immunogenic tumors to grow, although their relative contributions change with time. The evidence suggests that primary T1 tumors self-regulate their immune content depending on their size and this can influence the lymphoid compartment of T2 tumors, especially with respect to Tregs. Tumor burden is a major confounding factor in immune analysis that has to be taken into consideration in experimental models and in the clinic. hRT caused complete local regression of primary tumors, which was accompanied by heavy infiltration of CD8+ T cells activated to express IFN-&gamma, and PD-1, while certain myeloid populations diminished. In spite of this active infiltrate, primary hRT failed to generate the systemic conditions required to cause abscopal regression of unirradiated microscopic tumors unless PD-1 blockade, which on its own was ineffective, was added to the RT regimen. The combination further increased local and systemically activated CD8+ T cells, but few other changes. This study emphasizes the subtle interplay between the immune system and tumors as they grow and how difficult it is for local RT, which can generate a local immune response that may help with primary tumor regression, to overcome the systemic barriers that are generated so as to effect immune regression of even small abscopal lesions.
- Published
- 2020
- Full Text
- View/download PDF
31. Focal Irradiation and Systemic TGFβ Blockade in Metastatic Breast Cancer
- Author
-
Sylvia Adams, Lin Hwang, Josephine A. Ratikan, James Sayre, Silvia C. Formenti, John A. Glaspy, Carol Felix, Dörthe Schaue, Kym F. Faull, William H. McBride, Judith D. Goldberg, Xiaochun Li, Sara A. Hurvitz, Percy Lee, Sandra Demaria, Mike W. Xie, and Begoña Comin-Anduix
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,Gastroenterology ,chemistry.chemical_compound ,0302 clinical medicine ,Transforming Growth Factor beta ,Positron Emission Tomography Computed Tomography ,Monoclonal ,Medicine ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Humanized ,Cancer ,Hazard ratio ,Fresolimumab ,Abscopal effect ,Middle Aged ,Combined Modality Therapy ,Metastatic breast cancer ,6.5 Radiotherapy and other non-invasive therapies ,Treatment Outcome ,Oncology ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Female ,Drug Monitoring ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Breast Neoplasms ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,Article ,Antibodies ,03 medical and health sciences ,Clinical Research ,Median follow-up ,Internal medicine ,Breast Cancer ,Humans ,Oncology & Carcinogenesis ,Neoplasm Staging ,Aged ,Radiotherapy ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Survival Analysis ,Blockade ,Radiation therapy ,030104 developmental biology ,chemistry ,business ,Biomarkers - Abstract
Purpose: This study examined the feasibility, efficacy (abscopal effect), and immune effects of TGFβ blockade during radiotherapy in metastatic breast cancer patients. Experimental Design: Prospective randomized trial comparing two doses of TGFβ blocking antibody fresolimumab. Metastatic breast cancer patients with at least three distinct metastatic sites whose tumor had progressed after at least one line of therapy were randomized to receive 1 or 10 mg/kg of fresolimumab, every 3 weeks for five cycles, with focal radiotherapy to a metastatic site at week 1 (three doses of 7.5 Gy), that could be repeated to a second lesion at week 7. Research bloods were drawn at baseline, week 2, 5, and 15 to isolate PBMCs, plasma, and serum. Results: Twenty-three patients were randomized, median age 57 (range 35–77). Seven grade 3/4 adverse events occurred in 5 of 11 patients in the 1 mg/kg arm and in 2 of 12 patients in the 10 mg/kg arm, respectively. Response was limited to three stable disease. At a median follow up of 12 months, 20 of 23 patients are deceased. Patients receiving the 10 mg/kg had a significantly higher median overall survival than those receiving 1 mg/kg fresolimumab dose [hazard ratio: 2.73 with 95% confidence interval (CI), 1.02–7.30; P = 0.039]. The higher dose correlated with improved peripheral blood mononuclear cell counts and a striking boost in the CD8 central memory pool. Conclusions: TGFβ blockade during radiotherapy was feasible and well tolerated. Patients receiving the higher fresolimumab dose had a favorable systemic immune response and experienced longer median overall survival than the lower dose group. Clin Cancer Res; 24(11); 2493–504. ©2018 AACR.
- Published
- 2018
- Full Text
- View/download PDF
32. Biomarqueurs inflammatoires et immunologiques de réponse à la radiothérapie
- Author
-
J.P. Nesseler, Philippe Nickers, Dörthe Schaue, and William H. McBride
- Subjects
0301 basic medicine ,Tumor microenvironment ,Chemokine ,Tumor hypoxia ,biology ,business.industry ,medicine.medical_treatment ,Context (language use) ,Radiation therapy ,03 medical and health sciences ,Circulating biomarkers ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,FMISO - Abstract
In radiotherapy, the treatment is adapted to each individual to protect healthy tissues but delivers most of time a standard dose according to the tumor histology and site. The only biomarkers studied to individualize the treatment are the HPV status with radiation dose de-escalation strategies, and tumor hypoxia with dose escalation to hypoxic subvolumes using FMISO- or FAZA-PET imaging. In the last decades, evidence has grown about the contribution of the immune system to radiation tumor response. Many preclinical studies have identified some of the mechanisms involved. In this context, we have realised a systematic review to highlight potential inflammatory and immune biomarkers of radiotherapy response. Some are inside the tumor microenvironment, as lymphocyte infiltration or PD-L1 expression, others are circulating biomarkers, including different types of hematological cells, cytokines and chemokines.
- Published
- 2018
- Full Text
- View/download PDF
33. If It Seems Too Good to Be True…
- Author
-
Michael L. Steinberg, Erina Vlashi, William H. McBride, and Frank Pajonk
- Subjects
Cancer Research ,Radiation ,Text mining ,Oncology ,business.industry ,Medicine ,Radiology, Nuclear Medicine and imaging ,business ,Data science - Published
- 2019
- Full Text
- View/download PDF
34. Interleukin 32 expression in human melanoma
- Author
-
Dörthe Schaue, William H. McBride, Helicia Paz, Thomas G. Graeber, Anusha Kalbasi, James S. Economou, Deena M. Maurer, Jennifer Tsoi, Lisa H. Butterfield, Catherine S. Grasso, and Antoni Ribas
- Subjects
0301 basic medicine ,Skin Neoplasms ,Biopsy ,lcsh:Medicine ,Interleukin 32 (IL32) ,Medical and Health Sciences ,0302 clinical medicine ,Gene expression ,Transcriptional regulation ,Tumor Microenvironment ,2.1 Biological and endogenous factors ,Lymphocytes ,Aetiology ,Neoplasm Metastasis ,Melanoma ,Cancer ,Tumor ,General Medicine ,Microphthalmia-associated transcription factor ,3. Good health ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Biotechnology ,Myeloid polarization ,Melanoma dedifferentiation ,IL32 transcriptional regulation ,Immunology ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Proinflammatory cytokine ,Cell Line ,03 medical and health sciences ,Interferon-gamma ,Interleukin 32 ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,medicine ,Genetics ,Humans ,Tumor-Infiltrating ,Tumor microenvironment ,Neoplastic ,Tumor Necrosis Factor-alpha ,Research ,Gene Expression Profiling ,Interleukins ,Human Genome ,lcsh:R ,Gene signature ,Cell Dedifferentiation ,medicine.disease ,Microarray Analysis ,Immune infiltration ,030104 developmental biology ,Gene Expression Regulation ,Cancer research ,Transcriptome - Abstract
Background Various proinflammatory cytokines can be detected within the melanoma tumor microenvironment. Interleukin 32 (IL32) is produced by T cells, NK cells and monocytes/macrophages, but also by a subset of melanoma cells. We sought to better understand the biology of IL32 in human melanoma. Methods We analyzed RNA sequencing data from 53 in-house established human melanoma cell lines and 479 melanoma tumors from The Cancer Genome Atlas dataset. We evaluated global gene expression patterns associated with IL32 expression. We also evaluated the impact of proinflammatory molecules TNFα and IFNγ on IL32 expression and dedifferentiation in melanoma cell lines in vitro. In order to study the transcriptional regulation of IL32 in these cell lines, we cloned up to 10.5 kb of the 5′ upstream region of the human IL32 gene into a luciferase reporter vector. Results A significant proportion of established human melanoma cell lines express IL32, with its expression being highly correlated with a dedifferentiation genetic signature (high AXL/low MITF). Non IL32-expressing differentiated melanoma cell lines exposed to TNFα or IFNγ can be induced to express the three predominant isoforms (α, β, γ) of IL32. Cis-acting elements within this 5′ upstream region of the human IL32 gene appear to govern both induced and constitutive gene expression. In the tumor microenvironment, IL32 expression is highly correlated with genes related to T cell infiltration, and also positively correlates with high AXL/low MITF dedifferentiated gene signature. Conclusions Expression of IL32 in human melanoma can be induced by TNFα or IFNγ and correlates with a treatment-resistant dedifferentiated genetic signature. Constitutive and induced expression are regulated, in part, by cis-acting sequences within the 5′ upstream region. Electronic supplementary material The online version of this article (10.1186/s12967-019-1862-y) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
35. The tubercular badger and the uncertain curve:- The need for a multiple stressor approach in environmental radiation protection
- Author
-
Gibin G. Powathil, Brian Austin, David Copplestone, Nele Horemans, Jill Sutcliffe, William H. McBride, Jessica Goodman, Carmel Mothersill, Timothy A. Mousseau, François Bréchignac, Penny A. Jeggo, Anthony O'Hare, Paul N. Schofield, Colin Seymour, Rao V. Papineni, Michael Abend, Stanislav A. Geras’kin, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), Schofield, Paul [0000-0002-5111-7263], and Apollo - University of Cambridge Repository
- Subjects
medicine.medical_specialty ,010501 environmental sciences ,Radiation Dosage ,Radioecology ,01 natural sciences ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Multidisciplinary approach ,Radiation, Ionizing ,medicine ,Animals ,Dose effect ,030212 general & internal medicine ,Ecosystem approach ,0105 earth and related environmental sciences ,General Environmental Science ,Radiation protection ,Resilience ,business.industry ,Stressor ,Low dose ,Dose-Response Relationship, Radiation ,Radiation Exposure ,Scotland ,Risk analysis (engineering) ,[SDE]Environmental Sciences ,Ecosphere ,business ,Dose rate ,Psychology - Abstract
This article presents the results of a workshop held in Stirling, Scotland in June 2018, called to examine critically the effects of low-dose ionising radiation on the ecosphere. The meeting brought together participants from the fields of low- and high-dose radiobiology and those working in radioecology to discuss the effects that low doses of radiation have on non-human biota. In particular, the shape of the low-dose response relationship and the extent to which the effects of low-dose and chronic exposure may be predicted from high dose rate exposures were discussed. It was concluded that high dose effects were not predictive of low dose effects. It followed that the tools presently available were deemed insufficient to reliably predict risk of low dose exposures in ecosystems. The workshop participants agreed on three major recommendations for a path forward. First, as treating radiation as a single or unique stressor was considered insufficient, the development of a multidisciplinary approach is suggested to address key concerns about multiple stressors in the ecosphere. Second, agreed definitions are needed to deal with the multiplicity of factors determining outcome to low dose exposures as a term can have different meanings in different disciplines. Third, appropriate tools need to be developed to deal with the different time, space and organisation level scales. These recommendations permit a more accurate picture of prospective risks. © 2018 The Author(s)
- Published
- 2019
- Full Text
- View/download PDF
36. Plasticity of Myeloid Cells during Oral Barrier Wound Healing and the Development of Bisphosphonate-related Osteonecrosis of the Jaw
- Author
-
Ichiro Nishimura, Keiichi Kanayama, Akishige Hokugo, Kenzo Morinaga, William H. McBride, Sil Park, Kawaljit Kaur, Yujie Sun, Anahid Jewett, Anna Kozlowska, and Jun Li
- Subjects
0301 basic medicine ,Pathology ,Myeloid ,medicine.medical_treatment ,bone ,Medical and Health Sciences ,Biochemistry ,necrosis ,Mice ,0302 clinical medicine ,Bone Marrow ,Antigens, Ly ,2.1 Biological and endogenous factors ,Myeloid Cells ,Aetiology ,neutrophil ,Molecular Bases of Disease ,Anatomy ,Biological Sciences ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Bisphosphonate-Associated Osteonecrosis of the Jaw ,medicine.symptom ,Biochemistry & Molecular Biology ,medicine.medical_specialty ,bone marrow ,Lesion ,03 medical and health sciences ,Immune system ,medicine ,Animals ,Antigens ,Dental/Oral and Craniofacial Disease ,Molecular Biology ,Mouth ,Wound Healing ,Bisphosphonate-associated osteonecrosis of the jaw ,business.industry ,Inflammatory and immune system ,Cell Biology ,Bisphosphonate ,medicine.disease ,stomatognathic diseases ,030104 developmental biology ,Ly ,Tooth Extraction ,Chemical Sciences ,Bone marrow ,Wound healing ,Osteonecrosis of the jaw ,business - Abstract
Injury to the barrier tissue initiates a rapid distribution of myeloid immune cells from bone marrow, which guide sound wound healing. Bisphosphonates, a widely used anti-bone resorptive drug with minimal systemic side effects, have been linked to an abnormal wound healing in the oral barrier tissue leading to, in some cases, osteonecrosis of the jaw (ONJ). Here we report that the development of ONJ may involve abnormal phenotypic plasticity of Ly6G+/Gr1+ myeloid cells in the oral barrier tissue undergoing tooth extraction wound healing. A bolus intravenous zoledronate (ZOL) injection to female C57Bl/6 mice followed by maxillary first molar extraction resulted in the development of ONJ-like lesion during the second week of wound healing. The multiplex assay of dissociated oral barrier cells exhibited the secretion of cytokines and chemokines, which was significantly modulated in ZOL mice. Tooth extraction-induced distribution of Ly6G+/Gr1+ cells in the oral barrier tissue increased in ZOL mice at week 2. ONJ-like lesion in ZOL mice contained Ly6G+/Gr1+ cells with abnormal size and morphology as well as different flow cytometric staining intensity. When anti-Ly6G (Gr1) antibody was intraperitoneally injected for 5 days during the second week of tooth extraction, CD11b+GR1(hi) cells in bone marrow and Ly6G+ cells in the oral barrier tissue were depleted, and the development of ONJ-like lesion was significantly attenuated. This study suggests that local modulation of myeloid cell plasticity in the oral barrier tissue may provide the basis for pathogenesis and thus therapeutic as well as preventive strategy of ONJ.
- Published
- 2016
- Full Text
- View/download PDF
37. Defenses against Pro-oxidant Forces - Maintenance of Cellular and Genomic Integrity and Longevity
- Author
-
Razmik Mirzayans, David Murray, and William H. McBride
- Subjects
0301 basic medicine ,Genome instability ,Cell cycle checkpoint ,Lung Neoplasms ,Ionizing ,DNA Repair ,Ataxia Telangiectasia Mutated Proteins ,Mitochondrion ,medicine.disease_cause ,Medical and Health Sciences ,Antioxidants ,Carcinoma, Non-Small-Cell Lung ,Radiation, Ionizing ,DNA-(Apurinic or Apyrimidinic Site) Lyase ,2.1 Biological and endogenous factors ,Aetiology ,Non-Small-Cell Lung ,Radiation ,Radiation Exposure ,Biological Sciences ,Cell biology ,Physical Sciences ,Cyclin-Dependent Kinase Inhibitor p21 ,DNA damage ,DNA repair ,NF-E2-Related Factor 2 ,1.1 Normal biological development and functioning ,Longevity ,Biophysics ,Biology ,Article ,Genomic Instability ,Cell Physiological Phenomena ,03 medical and health sciences ,Underpinning research ,medicine ,Genetics ,Animals ,Humans ,Radiology, Nuclear Medicine and imaging ,Oncology & Carcinogenesis ,Transcription factor ,Cellular compartment ,Protein Processing ,Carcinoma ,Post-Translational ,Oxidative Stress ,030104 developmental biology ,Protein Biosynthesis ,Generic health relevance ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species ,Protein Processing, Post-Translational ,Oxidative stress ,DNA Damage - Abstract
There has been enormous recent progress in understanding how human cells respond to oxidative stress, such as that caused by exposure to ionizing radiation. We have witnessed a significant deciphering of the events that underlie how antioxidant responses counter pro-oxidant damage to key biological targets in all cellular compartments, including the genome and mitochondria. These cytoprotective responses include: 1. The basal cellular repertoire of antioxidant capabilities and its supporting cast of facilitator enzymes; and 2. The inducible phase of the antioxidant response, notably that mediated by the Nrf2 transcription factor. There has also been frenetic progress in defining how reactive electrophilic species swamp existing protective mechanisms to augment DNA damage, events that are embodied in the cellular "DNA-damage response", including cell cycle checkpoint activation and DNA repair, which occur on a time scale of hours to days, as well as the implementation of cellular responses such as apoptosis, autophagy, senescence and reprograming that extend the time period of damage sensing and response into weeks, months and years. It has become apparent that, in addition to the initial oxidative insult, cells typically undergo further waves of secondary reactive oxygen/nitrogen species generation, DNA damage and signaling and that these may reemerge long after the initial events have subsided, probably being driven, at least in part, by persisting DNA damage. These reactive oxygen/nitrogen species are an integral part of the pathological consequences of radiation exposure and may persist across multiple cell divisions. Because of the pervasive nature of oxidative stress, a cell will manifest different responses in different subcellular compartments and to different levels of stress injury. Aspects of these compartmentalized responses can involve the same proteins (such as ATM, p53 and p21) but in different functional guises, e.g., in cytoplasmic versus nuclear responses or in early- versus late-phase events. Many of these responses involve gene activation and new protein synthesis as well as a plethora of post-translational modifications of both basal and induced response proteins. It is these responses that we focus on in this review.
- Published
- 2018
38. 1-(4-nitrobenzenesulfonyl)-4-penylpiperazine increases the number of Peyer's patch-associated regenerating crypts in the small intestines after radiation injury
- Author
-
Mohammad Saki, Kruttika Bhat, Renuka Ramanathan, Paul Medina, Sara Duhachek-Muggy, Frank Pajonk, Dörthe Schaue, William H. McBride, Claudia Alli, Erina Vlashi, Julian P. Whitelegge, and Robert Damoiseaux
- Subjects
Crypt ,Apoptosis ,Radiation-Protective Agents ,Biology ,Piperazines ,Article ,030218 nuclear medicine & medical imaging ,Flow cytometry ,03 medical and health sciences ,Mice ,Peyer's Patches ,Random Allocation ,0302 clinical medicine ,Immune system ,Intestine, Small ,medicine ,Animals ,Regeneration ,Radiology, Nuclear Medicine and imaging ,Intestinal Mucosa ,Radiation Injuries ,Nitrobenzenes ,Mice, Inbred C3H ,medicine.diagnostic_test ,Peyer's patch ,Germinal center ,Hematology ,Total body irradiation ,Molecular biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Lymphatic system ,Oncology ,030220 oncology & carcinogenesis ,Female ,Whole-Body Irradiation - Abstract
OBJECTIVE: Exposure to lethal doses of radiation has severe effects on normal tissues. Exposed individuals experience a plethora of symptoms in different organ systems including the gastrointestinal (GI) tract, summarized as Acute Radiation Syndrome (ARS). There are currently no approved drugs for mitigating GI-ARS. A recent high-throughput screen performed at the UCLA Center for Medical Countermeasures against Radiation identified compounds containing sulfonylpiperazine groups with radiation mitigation properties to the hematopoietic system and the gut. Among these 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine (Compound #5) efficiently mitigated gastrointestinal ARS. However, the mechanism of action and target cells of this drug is still unknown. In this study we examined if Compound #5 affects gut-associated lymphoid tissue (GALT) with its subepithelial domes called Peyer’s patches. METHODS: C3H mice were irradiated with 0 or 12 Gy total body irradiation (TBI). A single dose of Compound #5 or solvent was administered subcutaneously 24 hours later. 48 hours after irradiation the mice were sacrificed, and the guts examined for changes in the number of visible Peyer’s patches. In some experiments the mice received 4 daily injections of treatment and were sacrificed 96 hours after TBI. For immune histochemistry gut tissues were fixed in formalin and embedded in paraffin blocks. Sections were stained with H&E, anti-Ki67 or a TUNEL assay to assess the number of regenerating crypts, mitotic and apoptotic indices. Cells isolated from Peyer’s patches were subjected to immune profiling using flow cytometry. RESULTS: Compound #5 significantly increased the number of visible Peyer’s patches when compared to its control in non-irradiated and irradiated mice. Additionally, assessment of total cells per Peyer’s patch isolated from these mice demonstrated an overall increase in the total number of Peyer’s patch cells per mouse in Compound #5-treated mice. In non-irradiated animals the number of CD11b(high) in Peyer’s patches increased significantly. These Compound #5-driven increases did not coincide with a decrease in apoptosis or an increase in proliferation in the germinal centers inside the Peyer’s patches 24 hours after drug treatment. A single dose of Compound #5 significantly increased the number of CD45(+) cells after 12 Gy TBI. Importantly, 96 hours after 12 Gy TBI Compound #5 induced a significant rise in the number of visible Peyer’s patches and the number of Peyer’s patch-associated regenerating crypts. CONCLUSION: In summary, our study provides evidence that Compound #5 leads to an influx of immune cells into GALT, thereby supporting crypt regeneration preferentially in the proximity of Peyer’s patches.
- Published
- 2018
39. Opportunities and challenges of radiotherapy for treating cancer
- Author
-
Dörthe Schaue and William H. McBride
- Subjects
medicine.medical_specialty ,Radiobiology ,Tumour heterogeneity ,medicine.medical_treatment ,Oncology and Carcinogenesis ,Translational research ,Host tissue ,Article ,Vaccine Related ,Neoplasms ,Humans ,Medicine ,Medical physics ,Dose Fractionation ,Cancer ,Radiation ,Radiotherapy ,business.industry ,Radiation dose ,Dose fractionation ,medicine.disease ,Radiation therapy ,5.5 Radiotherapy and other non-invasive therapies ,Oncology ,Immunization ,Dose Fractionation, Radiation ,Development of treatments and therapeutic interventions ,business - Abstract
The past 20 years have seen dramatic changes in the delivery of radiation therapy, but the impact of radiobiology on the clinic has been far less substantial. A major consideration in the use of radiotherapy has been on how best to exploit differences between the tumour and host tissue characteristics, which in the past has been achieved empirically by radiation-dose fractionation. New advances are uncovering some of the mechanistic processes that underlie this success story. In this Review, we focus on how these processes might be targeted to improve the outcome of radiotherapy at the individual patient level. This approach would seem a more productive avenue of treatment than simply trying to increase the radiation dose delivered to the tumour.
- Published
- 2015
- Full Text
- View/download PDF
40. Defined Sensing Mechanisms and Signaling Pathways Contribute to the Global Inflammatory Gene Expression Output Elicited by Ionizing Radiation
- Author
-
Ann-Jay Tong, William H. McBride, Prabhat Kumar Purbey, Keisuke S. Iwamoto, Philip O. Scumpia, Stephen T. Smale, and Peter J. Kim
- Subjects
0301 basic medicine ,p53 ,Ionizing ,Transcription, Genetic ,Receptor, Interferon alpha-beta ,DNA-Activated Protein Kinase ,Ataxia Telangiectasia Mutated Proteins ,p38 Mitogen-Activated Protein Kinases ,Interferon alpha-beta ,Mice ,Gene Knockout Techniques ,Interferon ,Radiation, Ionizing ,Transcriptional regulation ,Immunology and Allergy ,Cluster Analysis ,2.1 Biological and endogenous factors ,Inducer ,Aetiology ,Extracellular Signal-Regulated MAP Kinases ,Radiation ,Kinase ,Adaptor Proteins ,Innate Immunity ,Cell biology ,Infectious Diseases ,Signal transduction ,medicine.symptom ,Transcription ,medicine.drug ,Signal Transduction ,Receptor ,Transcriptional Activation ,Interferon Response ,NF-E2-Related Factor 2 ,Immunology ,Inflammation ,Biology ,Article ,Dose-Response Relationship ,03 medical and health sciences ,Genetic ,Transcriptional Regulator ERG ,medicine ,Genetics ,Animals ,Humans ,Gene ,Transcriptional Regulation ,Innate immune system ,Macrophages ,Gene Expression Profiling ,Inflammatory and immune system ,Membrane Proteins ,Dose-Response Relationship, Radiation ,Vesicular Transport ,Adaptor Proteins, Vesicular Transport ,030104 developmental biology ,Gene Expression Regulation ,ATM ,Myeloid Differentiation Factor 88 ,Ionizing Radiation ,Interferons ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species - Abstract
Summary Environmental insults are often detected by multiple sensors that activate diverse signaling pathways and transcriptional regulators, leading to a tailored transcriptional output. To understand how a tailored response is coordinated, we examined the inflammatory response elicited in mouse macrophages by ionizing radiation (IR). RNA-sequencing studies revealed that most radiation-induced genes were strongly dependent on only one of a small number of sensors and signaling pathways, notably the DNA damage-induced kinase ATM, which regulated many IR-response genes, including interferon response genes, via an atypical IRF1-dependent, STING-independent mechanism. Moreover, small, defined sets of genes activated by p53 and NRF2 accounted for the selective response to radiation in comparison to a microbial inducer of inflammation. Our findings reveal that genes comprising an environmental response are activated by defined sensing mechanisms with a high degree of selectivity, and they identify distinct components of the radiation response that might be susceptible to therapeutic perturbation.
- Published
- 2017
41. National Institutes of Health Funding in Radiation Oncology: A Snapshot
- Author
-
Frank Pajonk, Michael L. Steinberg, Erina Vlashi, and William H. McBride
- Subjects
Cancer Research ,medicine.medical_specialty ,Medical education ,Radiation ,Descriptive statistics ,business.industry ,education ,Alternative medicine ,MEDLINE ,Work force ,Small field ,Fiscal year ,Oncology ,Radiation oncology ,medicine ,Radiology, Nuclear Medicine and imaging ,business ,health care economics and organizations ,Health funding - Abstract
Currently, pay lines for National Institutes of Health (NIH) grants are at a historical low. In this climate of fierce competition, knowledge about the funding situation in a small field like radiation oncology becomes very important for career planning and recruitment of faculty. Unfortunately, these data cannot be easily extracted from the NIH's database because it does not discriminate between radiology and radiation oncology departments. At the start of fiscal year 2013 we extracted records for 952 individual grants, which were active at the time of analysis from the NIH database. Proposals originating from radiation oncology departments were identified manually. Descriptive statistics were generated using the JMP statistical software package. Our analysis identified 197 grants in radiation oncology. These proposals came from 134 individual investigators in 43 academic institutions. The majority of the grants (118) were awarded to principal investigators at the full professor level, and 122 principal investigators held a PhD degree. In 79% of the grants, the research topic fell into the field of biology, 13% in the field of medical physics. Only 7.6% of the proposals were clinical investigations. Our data suggest that the field of radiation oncology is underfunded by the NIH and that the current level of support does not match the relevance of radiation oncology for cancer patients or the potential of its academic work force.
- Published
- 2013
- Full Text
- View/download PDF
42. Cutaneous wound healing through paradoxical MAPK activation by BRAF inhibitors
- Author
-
Mohammad Atefi, Rong Rong Huang, Begoña Comin-Anduix, Felipe Meira de-Faria, Ariel M. Azhdam, Jing Jiao, Roger S. Lo, Michael W. Xie, Tatiana Segura, Helena Escuin-Ordinas, Robert L. Modlin, Alistair J. Cochran, Paige Krystofinski, Willy Hugo, Harvey R. Herschman, Shuoran Li, Antoni Ribas, Sara Marie D. Komenan, Susan Realegeno, William H. McBride, Matteo Pellegrini, and Lu Sun
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Keratinocytes ,Indoles ,Skin Neoplasms ,endocrine system diseases ,Carcinogenesis ,Administration, Topical ,General Physics and Astronomy ,Mitogen-activated protein kinase kinase ,Mice ,Neoplasms ,Medicine ,Vemurafenib ,Inbred BALB C ,Cancer ,Skin ,Mice, Inbred BALB C ,Sulfonamides ,Multidisciplinary ,Tumor ,integumentary system ,Kinase ,MEK inhibitor ,Melanoma ,Incidence ,Cell Cycle ,3. Good health ,Treatment Outcome ,Topical ,5.1 Pharmaceuticals ,Administration ,Carcinoma, Squamous Cell ,Female ,Development of treatments and therapeutic interventions ,Mitogen-Activated Protein Kinases ,medicine.drug ,Proto-Oncogene Proteins B-raf ,MAP Kinase Signaling System ,Pyridones ,Science ,Pyrimidinones ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Experimental ,Cell Line, Tumor ,Animals ,Humans ,Protein kinase A ,neoplasms ,Protein Kinase Inhibitors ,Mitogen-Activated Protein Kinase Kinases ,Wound Healing ,business.industry ,Carcinoma ,General Chemistry ,Neoplasms, Experimental ,medicine.disease ,digestive system diseases ,030104 developmental biology ,Squamous Cell ,Cancer research ,Carcinogens ,Wound healing ,business - Abstract
BRAF inhibitors are highly effective therapies for the treatment of BRAFV600-mutated melanoma, with the main toxicity being a variety of hyperproliferative skin conditions due to paradoxical activation of the mitogen-activated protein kinase (MAPK) pathway in BRAF wild-type cells. Most of these hyperproliferative skin changes improve when a MEK inhibitor is co-administered, as it blocks paradoxical MAPK activation. Here we show how the BRAF inhibitor vemurafenib accelerates skin wound healing by inducing the proliferation and migration of human keratinocytes through extracellular signal-regulated kinase (ERK) phosphorylation and cell cycle progression. Topical treatment with vemurafenib in two wound-healing mice models accelerates cutaneous wound healing through paradoxical MAPK activation; addition of a mitogen-activated protein kinase kinase (MEK) inhibitor reverses the benefit of vemurafenib-accelerated wound healing. The same dosing regimen of topical BRAF inhibitor does not increase the incidence of cutaneous squamous cell carcinomas in mice. Therefore, topical BRAF inhibitors may have clinical applications in accelerating the healing of skin wounds., BRAF inhibitors often show skin-hyperproliferative side effects in melanoma patients. Here, the authors demonstrate that BRAF inhibitors can be used to enhance skin wound healing through the MAPK- ERK pathway activation that positively regulates the proliferation of keratinocytes.
- Published
- 2016
- Full Text
- View/download PDF
43. Spinal Cord Tolerance to Reirradiation With Single-Fraction Radiosurgery: A Swine Model
- Author
-
Paul M. Medin, William H. McBride, Timothy D. Solberg, Albert J. van der Kogel, James Sayre, and Ryan D. Foster
- Subjects
Cancer Research ,Time Factors ,Swine ,medicine.medical_treatment ,Infarction ,Radiation Dosage ,Radiosurgery ,Radiation Tolerance ,Article ,Translational research [ONCOL 3] ,Paralysis ,medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,Gait Disorders, Neurologic ,Myelin Sheath ,Radiation ,business.industry ,Dose-Response Relationship, Radiation ,Histology ,Spinal cord ,medicine.disease ,Radiation therapy ,Radiation Injuries, Experimental ,Dose–response relationship ,medicine.anatomical_structure ,Spinal Cord ,Oncology ,Retreatment ,Cervical Vertebrae ,Swine, Miniature ,Female ,medicine.symptom ,Nuclear medicine ,business ,Cervical vertebrae - Abstract
Item does not contain fulltext PURPOSE: This study was performed to determine swine spinal cord tolerance to single-fraction, partial-volume irradiation 1 year after receiving uniform irradiation to 30 Gy in 10 fractions. METHODS AND MATERIALS: A 10-cm length of spinal cord (C3-T1) was uniformly irradiated to 30 Gy in 10 consecutive fractions and reirradiated 1 year later with a single radiosurgery dose centered within the previously irradiated segment. Radiosurgery was delivered to a cylindrical volume approximately 5 cm in length and 2 cm in diameter, which was positioned laterally to the cervical spinal cord, resulting in a dose distribution with the 90%, 50%, and 10% isodose lines traversing the ipsilateral, central, and contralateral spinal cord, respectively. Twenty-three pigs were stratified into six dose groups with mean maximum spinal cord doses of 14.9 +/- 0.1 Gy (n = 2), 17.1 +/- 0.3 Gy (n = 3), 19.0 +/- 0.1 Gy (n = 5), 21.2 +/- 0.1 Gy (n = 5), 23.4 +/- 0.2 Gy (n = 5), and 25.4 +/- 0.4 Gy (n = 3). The mean percentage of spinal cord volumes receiving >/=10 Gy for the same groups were 34% +/- 1%, 40% +/- 1%, 46% +/- 3%, 52% +/- 1%, 56 +/- 3%, and 57% +/- 1%. The study endpoint was motor neurologic deficit as determined by a change in gait during a 1- year follow-up period. RESULTS: A steep dose-response curve was observed with a 50% incidence of paralysis (ED(50)) for the maximum point dose of 19.7 Gy (95% confidence interval, 17.4-21.4). With two exceptions, histology was unremarkable in animals with normal neurologic status, while all animals with motor deficits showed some degree of demyelination and focal white matter necrosis on the irradiated side, with relative sparing of gray matter. Histologic comparison with a companion study of de novo irradiated animals revealed that retreatment responders had more extensive tissue damage, including infarction of gray matter, only at prescription doses >20 Gy. CONCLUSION: Pigs receiving spinal radiosurgery 1 year after receiving 30 Gy in 10 fractions were not at significantly higher risk of developing motor deficits than pigs that received radiosurgery alone.
- Published
- 2012
- Full Text
- View/download PDF
44. 5-Aminoimidazole-4-Carboxamide Riboside Enhances Effect of Ionizing Radiation in PC3 Prostate Cancer Cells
- Author
-
Sofie Isebaert, Karin Haustermans, William H. McBride, Johannes V. Swinnen, and Adrian C. Begg
- Subjects
Male ,Adenosine monophosphate ,Radiation-Sensitizing Agents ,Cancer Research ,Small interfering RNA ,medicine.medical_specialty ,Time Factors ,Cell Survival ,DNA repair ,Apoptosis ,AMP-Activated Protein Kinases ,Radiation Tolerance ,chemistry.chemical_compound ,AMP-activated protein kinase ,Cell Line, Tumor ,Radiation, Ionizing ,Internal medicine ,Humans ,Medicine ,Radiology, Nuclear Medicine and imaging ,Viability assay ,Radiosensitivity ,RNA, Small Interfering ,Tumor Stem Cell Assay ,Cell Proliferation ,Radiation ,biology ,Cell growth ,business.industry ,Prostatic Neoplasms ,AMPK ,Ribonucleotides ,Aminoimidazole Carboxamide ,Enzyme Activation ,Metabolism ,Endocrinology ,Oncology ,chemistry ,Gene Knockdown Techniques ,biology.protein ,Cancer research ,Ribonucleosides ,Reactive Oxygen Species ,business - Abstract
Purpose The nucleoside 5-aminoimidazole-4-carboxamide riboside (AICAR) is a low-energy mimetic and adenosine monophosphate (AMP)-activated protein kinase (AMPK) agonist that can affect the phenotype of malignant cells by diminishing their anabolism. It does this by being converted to 5-aminoimidazole-4-carboxamide ribotide (ZMP), an AMP analog. We combined this promising antineoplastic agent with ionizing radiation in an attempt to increase its efficacy. Methods and Materials The effect of AICAR on cell proliferation, cell viability, apoptosis, reactive oxygen species production, radiosensitivity, and AMPK activation was determined in the human prostate cancer cell line PC3. To elucidate the radiosensitizing mechanism, clonogenic survival assays in the presence of a drug agonist or antagonist or with small interfering RNA targeting AMPK were done, as well as measurements of ZMP production and double strand break repair. Moreover, immunoblot analysis of the radiation response signaling pathways after AICAR treatment was performed. Results The incubation of human PC3 prostate cancer cells with AICAR-activated AMPK inhibited cell proliferation, decreased viability, increased apoptosis, and generated reactive oxygen species in a dose- and time-dependent manner. None of these endpoints gave more than additive effects when radiation was added. Radiosensitization was observed but only after 72 hours of treatment with 250 μM AICAR, suggesting that it was independent of AMPK activation. This finding was confirmed by small interfering RNA knockdown of AMPK. The mechanism of radiosensitization was associated with imbalanced deoxynucleotide pools owing to ZMP accumulation after AICAR administration that interfered with DNA repair. Conclusions Our findings on the favorable interaction between low doses of AICAR and ionizing radiation in PC3 cells could open new perspectives for the clinical use of this or similar compounds. However, additional research is still required to establish the ZMP pathway as being of general applicability.
- Published
- 2011
- Full Text
- View/download PDF
45. Radiation Enhances Regulatory T Cell Representation
- Author
-
James S. Economou, Dörthe Schaue, William H. McBride, Keisuke S. Iwamoto, John J. DeMarco, Yu-Pei Liao, Evelyn L. Kachikwu, and Nzhde Agazaryan
- Subjects
CD4-Positive T-Lymphocytes ,Male ,Cancer Research ,Regulatory T cell ,medicine.medical_treatment ,Population ,Mice, Transgenic ,chemical and pharmacologic phenomena ,Adenocarcinoma ,Radiation Tolerance ,T-Lymphocytes, Regulatory ,Article ,Mice ,Immune system ,Cell Line, Tumor ,medicine ,Animals ,Scattering, Radiation ,Cytotoxic T cell ,Radiology, Nuclear Medicine and imaging ,Lymphocyte Count ,IL-2 receptor ,education ,education.field_of_study ,Radiation ,business.industry ,Interleukin-2 Receptor alpha Subunit ,CD24 Antigen ,Prostatic Neoplasms ,FOXP3 ,Dose-Response Relationship, Radiation ,Forkhead Transcription Factors ,hemic and immune systems ,Immunotherapy ,Hindlimb ,Mice, Inbred C57BL ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Immunology ,Female ,Lymph Nodes ,business ,Biomarkers ,Spleen ,Whole-Body Irradiation - Abstract
Purpose Immunotherapy could be a useful adjunct to standard cytotoxic therapies such as radiation in patients with micrometastatic disease, although successful integration of immunotherapy into treatment protocols will require further understanding of how standard therapies affect the generation of antitumor immune responses. This study was undertaken to evaluate the impact of radiation therapy (RT) on immunosuppressive T regulatory (Treg) cells. Methods and Materials Treg cells were identified as a CD4 + CD25 hi Foxp3 + lymphocyte subset, and their fate was followed in a murine TRAMP C1 model of prostate cancer in mice with and without RT. Results CD4 + CD25 hi Foxp3 + Treg cells increased in immune organs after local leg or whole-body radiation. A large part, but not all, of this increase after leg-only irradiation could be ascribed to radiation scatter and Treg cells being intrinsically more radiation resistant than other lymphocyte subpopulations, resulting in their selection. Their functional activity on a per-cell basis was not affected by radiation exposure. Similar findings were made with mice receiving local RT to murine prostate tumors growing in the leg. The importance of the Treg cell population in the response to RT was shown by systemic elimination of Treg cells, which greatly enhanced radiation-induced tumor regression. Conclusions We conclude that Treg cells are more resistant to radiation than other lymphocytes, resulting in their preferential increase. Treg cells may form an important homeostatic mechanism for tissues injured by radiation, and in a tumor context, they may assist in immune evasion during therapy. Targeting this population may allow enhancement of radiotherapeutic benefit through immune modulation.
- Published
- 2011
- Full Text
- View/download PDF
46. Insulin-Like Growth Factor–Type 1 Receptor Inhibitor NVP-AEW541 Enhances Radiosensitivity of PTEN Wild-Type but Not PTEN-Deficient Human Prostate Cancer Cells
- Author
-
Johannes V. Swinnen, Sofie Isebaert, William H. McBride, and Karin Haustermans
- Subjects
Male ,Radiation-Sensitizing Agents ,Cancer Research ,DNA Repair ,Cell Survival ,Down-Regulation ,Radiation Tolerance ,Receptor, IGF Type 1 ,DU145 ,Cell Line, Tumor ,Radioresistance ,Humans ,Medicine ,PTEN ,DNA Breaks, Double-Stranded ,Pyrroles ,Radiology, Nuclear Medicine and imaging ,Protein Kinase Inhibitors ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Radiation ,biology ,Cell growth ,business.industry ,Cell Cycle ,PTEN Phosphohydrolase ,Prostatic Neoplasms ,Cell cycle ,Neoplasm Proteins ,Pyrimidines ,Oncology ,Cancer cell ,Cancer research ,biology.protein ,business ,Proto-Oncogene Proteins c-akt - Abstract
Purpose During the past decade, many clinical trials with both monoclonal antibodies and small molecules that target the insulin-like growth factor–type 1 receptor (IGF-1R) have been launched. Despite the important role of IGF-1R signaling in radioresistance, studies of such agents in combination with radiotherapy are lagging behind. Therefore, the aim of this study was to investigate the effect of the small molecule IGF-1R kinase inhibitor NVP-AEW541 on the intrinsic radioresistance of prostate cancer cells. Methods and Materials The effect of NVP-AEW541 on cell proliferation, cell viability, IGF-1R signaling, radiosensitivity, cell cycle distribution, and double strand break repair was determined in three human prostate cancer cell lines (PC3, DU145, 22Rv1). Moreover, the importance of the PTEN pathway status was explored by means of transfection experiments with constitutively active Akt or inactive kinase-dead Akt. Results NVP-AEW541 inhibited cell proliferation and decreased cell viability in a time-and dose-dependent manner in all three cell lines. Radiosensitization was observed in the PTEN wild-type cell lines DU145 and 22Rv1 but not in the PTEN-deficient PC3 cell line. NVP-AEW541–induced radiosensitization coincided with downregulation of phospho-Akt levels and high levels of residual double strand breaks. The importance of PTEN status in the radiosensitization effect was confirmed by transfection experiments with constitutively active Akt or inactive kinase-dead Akt. Conclusions NVP-AEW541 enhances the effect of ionizing radiation in PTEN wild-type, but not in PTEN-deficient, prostate cancer cells. Proper patient selection based on the PTEN status of the tumor will be critical to the achievement of optimal results in clinical trials in which the combination of radiotherapy and this IGF-1R inhibitor is being explored.
- Published
- 2011
- Full Text
- View/download PDF
47. Small Azurin Derived Peptide Targets Ephrin Receptors for Radiotherapy
- Author
-
Hai Luong, Chun-Ling Jung, Piotr Ruchala, Ewa D. Micewicz, Dörthe Schaue, and William H. McBride
- Subjects
chemistry.chemical_classification ,Radiosensitizer ,Nicotinamide ,Chemistry ,Erythropoietin-producing hepatocellular (Eph) receptor ,Lewis lung carcinoma ,Bioengineering ,Peptide ,EPH receptor A2 ,Biochemistry ,Analytical Chemistry ,chemistry.chemical_compound ,Drug Discovery ,Cancer research ,Molecular Medicine ,Ephrin ,Receptor - Abstract
Many lung cancer treatment regimens include radiotherapy. We sought to improve the efficacy of such treatment by invoking the targeted delivery of a model radiosensitizer (nicotinamide) to malignant tissues. Ephrin receptors (Eph), which are often overexpressed in lung cancers, were selected as the target of our delivery system. Molecular targeting was achieved utilizing a small peptide derived from the C-terminal portion of azurin, a copper-containing redox protein (“cupredoxin”) that is capable of binding to ephrin receptors. We prepared and screened a sub-library of peptides derived from the C-terminal region of azurin and found several small analogues that bound ephrin receptors EphA2, EphB2, and EphB4. One such peptide, termed AzV36, was selected for conjugation with nicotinic acid via an amide bond to form AzV36-NicL. The resulting linear peptide contains 15 residues (including unusual and d-amino acids), is very stable in human serum, and can be easily manufactured. AzV36-NicL conjugate was tested in vivo for its ability to radiosensitize Lewis lung carcinoma (LCC) in artificial metastasis and solid tumor engraftment models. The compound increased the efficacy of radiotherapy with tumor colonies being ~2–13 fold lower than with radiation alone depending on experimental schedule. In contrast, equimolar amounts of unconjugated peptide (AzV36-L) or nicotinamide alone only marginally improved radiation efficacy. The targeted delivery of radiosensitizer(s) to ephrin receptors may enhance the efficacy of radiation treatment of lung cancer and of other cancers that overexpress ephrin receptor(s).
- Published
- 2011
- Full Text
- View/download PDF
48. Fibrin Microbeads Loaded with Mesenchymal Cells Support Their Long-Term Survival While Sealed at Room Temperature
- Author
-
Elena Gaberman, William H. McBride, Lilia Levdansky, Raphael Gorodetsky, Esther Lubzens, and Olga Gurevitch
- Subjects
Male ,Cell type ,Time Factors ,Cell Survival ,Cell ,Cell Culture Techniques ,Biomedical Engineering ,Medicine (miscellaneous) ,Bioengineering ,Cell Separation ,Biology ,Article ,Fibrin ,Cell therapy ,Young Adult ,medicine ,Humans ,Progenitor cell ,Cells, Cultured ,Cell survival ,Mesenchymal stem cell ,Infant, Newborn ,Temperature ,Mesenchymal Stem Cells ,Fibroblasts ,Hypoxia-Inducible Factor 1, alpha Subunit ,Microspheres ,Cell biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Cell culture ,biology.protein ,Biomedical engineering - Abstract
Efficient transfer of progenitor cells without affecting their survival is a key factor in any practical cell therapy. Fibrin microbeads (FMB) were developed as hard biodegradable cell carriers. The FMB could efficiently isolate mesenchymal stem cells (MSCs) from different sources and support the expansion of matrix-dependent cell types in a three-dimensional culture in slow rotation. The cells on FMB could also undergo induced differentiation for their eventual implantation to enhance tissue regeneration. FMB loaded with isolated human MSC (hMSC) were sealed in tubes topped up with medium. Almost full cell survival was recorded when the sealed cells were maintained in room temperature for up to 10 days, followed by a recovery period of 24 hrs at optimal conditions. Assay of cells recovery after such long room temperature storage showed ∼80%–100% survival of the cells on FMB, with only a marginal survival of cells that were kept in suspension without FMB in the same conditions. The hMSC that survived storage at room temperature preserved their profile of mesenchymal cell surface markers, their rate of proliferation, and their differentiation potential. The cell protective effect was not dependent on the presence of serum in the storage medium. It was clearly shown that over-expression of hypoxia induced factor-1α in hMSC with time, which may have protected the sealed cells on FMB at room temperature storage, was not necessarily related to extreme hypoxic stress. Foreskin normal fibroblasts on FMB sealed at room temperature were similarly protected, but with no elevation of their hypoxia-induced factor-1α expression. The results also show that FMB, unlike other commercially available cell carriers, could be used for delivery and shipping of progenitor cells at room temperature for extended time intervals. This could be highly useful for cell transfer for therapeutic application and for simplified cell transfer between different research centers.
- Published
- 2011
- Full Text
- View/download PDF
49. Radioprotective Effects of Bmi-1 Involve Epigenetic Silencing of Oxidase Genes and Enhanced DNA Repair in Normal Human Keratinocytes
- Author
-
No-Hee Park, Mo K. Kang, Qinghua Dong, Reuben H. Kim, Ki-Hyuk Shin, William H. McBride, Roy Y Kim, Ju-Eun Oh, and Wei Chen
- Subjects
Keratinocytes ,Senescence ,Jumonji Domain-Containing Histone Demethylases ,DNA Repair ,DNA repair ,DNA damage ,Dermatology ,Biochemistry ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Proto-Oncogene Proteins ,medicine ,Humans ,Epigenetics ,Radiation Injuries ,Molecular Biology ,Cells, Cultured ,Cellular Senescence ,030304 developmental biology ,Polycomb Repressive Complex 1 ,chemistry.chemical_classification ,0303 health sciences ,Oxidase test ,Reactive oxygen species ,biology ,Nuclear Proteins ,Cell Biology ,Molecular biology ,Repressor Proteins ,medicine.anatomical_structure ,Histone ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Oxidoreductases ,Reactive Oxygen Species ,Keratinocyte - Abstract
Normal human keratinocytes (NHKs) undergo premature senescence following exposure to ionizing radiation (IR). This study investigates the effect of Bmi-1, a polycomb group protein, on radiation-induced senescence response. When exposed to IR, NHK transduced with Bmi-1 (NHK/Bmi-1) showed reduced senescent phenotype and enhanced proliferation compared with control cells (NHK/B0). To investigate the underlying mechanism, we determined the production of reactive oxygen species (ROS), expression of ROS-generating enzymes, and DNA repair activities in cells. ROS level was increased upon irradiation but notably reduced by Bmi-1 transduction. Irradiation led to strong induction of oxidase genes, e.g., Lpo (lactoperoxidase), p22-phox, p47-phox, and Gp91, in NHK/B0 but their expression was almost completely silenced in NHK/Bmi-1. Induction of oxidase genes upon irradiation was linked with loss of trimethylated histone 3 at lysine 27 (H3K27Me3), but NHK/Bmi-1 expressed a higher level of H3K27Me3 compared with NHK/B0. Bmi-1 transduction suppressed IR-associated induction of jumanji domain containing 3 while enhancing the expression of EZH2, thereby preventing the loss of H3K27Me3 in the irradiated cells. Furthermore, NHK/Bmi-1 demonstrated increased repair of IR-induced DNA damage compared with NHK/B0. These results indicate that Bmi-1 elicits radioprotective effects on NHK by mitigating the genotoxicity of IR through epigenetic mechanisms.
- Published
- 2011
- Full Text
- View/download PDF
50. After the bomb drops: A new look at radiation-induced multiple organ dysfunction syndrome (MODS)
- Author
-
William H. McBride and Jacqueline P. Williams
- Subjects
Gastrointestinal tract ,medicine.medical_specialty ,Lung ,Radiological and Ultrasound Technology ,business.industry ,Multiple Organ Failure ,Acute Radiation Syndrome ,Context (language use) ,Radiation induced ,Comorbidity ,Total body irradiation ,medicine.disease ,Risk Assessment ,Article ,medicine.anatomical_structure ,Radiological weapon ,Prevalence ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Multiple organ dysfunction syndrome ,Intensive care medicine ,business ,Nuclear Warfare - Abstract
There is increasing concern that, since the Cold War era, there has been little progress regarding the availability of medical countermeasures in the event of either a radiological or nuclear incident. Fortunately, since much is known about the acute consequences that are likely to be experienced by an exposed population, the probability of survival from the immediate hematological crises after total body irradiation (TBI) has improved in recent years. Therefore focus has begun to shift towards later down-stream effects, seen in such organs as the gastrointestinal tract (GI), skin, and lung. However, the mechanisms underlying therapy-related normal tissue late effects, resulting from localised irradiation, have remained somewhat elusive and even less is known about the development of the delayed syndrome seen in the context of whole body exposures, when it is likely that systemic perturbations may alter tissue microenvironments and homeostasis.The sequence of organ failures observed after near-lethal TBI doses are similar in many ways to that of multiple organ dysfunction syndrome (MODS), leading to multiple organ failure (MOF). In this review, we compare the mechanistic pathways that underlie both MODS and delayed normal tissue effects since these may impact on strategies to identify radiation countermeasures.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.