1
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
2
|
Dutta A, Dahiya A. Quercetin 3-O rutinoside prevents gastrointestinal injury through regulation of apoptosis in 7.5 Gy total body irradiated mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 112:154692. [PMID: 36863087 DOI: 10.1016/j.phymed.2023.154692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 01/02/2023] [Accepted: 02/02/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Despite the heightened threat to unforeseen nuclear/radiological exposures worldwide, no countermeasures are currently approved to prevent gastrointestinal (GI) toxicity induced by radiation in humans. PURPOSE In this study, we aim to establish the gastroprotective role of flavonoid, Quercetin-3-O-rutinoside (Q-3-R) against 7.5 Gy total body gamma radiation dose that contributes to the hematopoietic syndrome. METHODS Q-3-R (10 mg/kg body weight) was administered intramuscularly to C57BL/6 male mice before exposure to 7.5 Gy and monitored for morbidity and mortality. The GI protection against radiation was ascertained by histopathological and xylose absorption studies. Intestinal apoptosis, crypt proliferation and apoptotic signaling were also investigated in different treatment groups. RESULTS We found that Q-3-R prevented the radiation-induced loss of mitochondrial membrane potential, maintained ATP levels, regulated the apoptotic pathway, and activated crypt cell proliferation in the intestine. Radiation-induced villi and crypt damage as well as mal-absorption were significantly minimized in the Q-3-R treated group. We observed 100% survival post Q-3-R administration against 33.3% lethality in 7.5 Gy (LD33.3/30) exposed C57BL/6 mice. The Q-3-R pre-treated mice that survived the 7.5 Gy dose revealed no pathological changes related to the development of fibrosis in the intestine and thickened mucosal wall till 4 months post irradiation. Complete hematopoietic recovery was observed in these surviving mice when compared to age matched control. CONCLUSION The findings revealed that Q-3-R regulated the apoptotic process to achieve GI protection against LD33.3/30 dose (7.5 Gy) that primarily caused death due to hematopoietic failure. The recovery observed in mice survivors suggested that this molecule may also have the potential to minimize side effects on normal tissues during radiotherapy.
Collapse
Affiliation(s)
- Ajaswrata Dutta
- Division of CBRN, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K Mazumdar Marg, Timarpur, Delhi 110054, India.
| | - Akshu Dahiya
- Division of CBRN, Institute of Nuclear Medicine and Allied Sciences (INMAS), Defence Research and Development Organization (DRDO), Brig. S.K Mazumdar Marg, Timarpur, Delhi 110054, India
| |
Collapse
|
3
|
Collao N, Sanders O, Caminiti T, Messeiller L, De Lisio M. Resistance and endurance exercise training improves muscle mass and the inflammatory/fibrotic transcriptome in a rhabdomyosarcoma model. J Cachexia Sarcopenia Muscle 2023; 14:781-793. [PMID: 36797054 PMCID: PMC10067492 DOI: 10.1002/jcsm.13185] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Rhabdomyosarcoma (RMS) is an aggressive soft tissue sarcoma that most often develops in children. Chemoradiation therapy is a standard treatment modality; however, the detrimental long-term skeletal muscle consequences of this therapy in juvenile cancer survivors include muscle atrophy and fibrosis resulting in decreased physical performance. Using a novel model of murine resistance and endurance exercise training, we investigate its role in preventing the long-term effects of juvenile RMS plus therapy. METHODS Four-week-old male (n = 10) and female (n = 10) C57Bl/6J mice were injected with M3-9-M RMS cell into the left gastrocnemius with the right limb serving as an internal control (CON). Mice received a systemic vincristine injection and then five doses of 4.8 Gy of gamma radiation localized to the left hindlimb (RMS + Tx). Mice were then randomly divided into either sedentary (SED) or resistance and endurance exercise training (RET) groups. Changes in exercise performance, body composition, myocellular adaptations and the inflammatory/fibrotic transcriptome were assessed. RESULTS RET improved endurance performance (P < 0.0001) and body composition (P = 0.0004) compared to SED. RMS + Tx resulted in significantly lower muscle weight (P = 0.015) and significantly smaller myofibre cross-sectional area (CSA) (P = 0.014). Conversely, RET resulted in significantly higher muscle weight (P = 0.030) and significantly larger Type IIA (P = 0.014) and IIB (P = 0.015) fibre CSA. RMS + Tx resulted in significantly more muscle fibrosis (P = 0.028), which was not prevented by RET. RMS + Tx resulted in significantly fewer mononuclear cells (P < 0.05) and muscle satellite (stem) cells (MuSCs) (P < 0.05) and significantly more immune cells (P < 0.05) than CON. RET resulted in significantly more fibro-adipogenic progenitors (P < 0.05), a trend for more MuSCs (P = 0.076) than SED and significantly more endothelial cells specifically in the RMS + Tx limb. Transcriptomic changes revealed significantly higher expression of inflammatory and fibrotic genes in RMS + Tx, which was prevented by RET. In the RMS + Tx model, RET also significantly altered expression of genes involved in extracellular matrix turnover. CONCLUSIONS Our study suggests that RET preserves muscle mass and performance in a model of juvenile RMS survivorship while partially restoring cellular dynamics and the inflammatory and fibrotic transcriptome.
Collapse
Affiliation(s)
- Nicolas Collao
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Olivia Sanders
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Taylor Caminiti
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Messeiller
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada
| | - Michael De Lisio
- School of Human Kinetics, University of Ottawa, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, Regenerative Medicine Program, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Wang X, Yuan R, Miao L, Li X, Guo Y, Tian H. Protective mechanism of a novel aminothiol compound on radiation-induced intestinal injury. Int J Radiat Biol 2023; 99:259-269. [PMID: 35583501 DOI: 10.1080/09553002.2022.2074163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE With the development of nuclear technology and radiotherapy, the risk of radiation injury has been increasing. Therefore, it is important to find an effective radiation-protective agent. In this study, we designed and synthesized a novel compound called compound 8, of which the radioprotective effect and mechanism were studied. MATERIALS AND METHODS Before being exposed to ionizing radiation, mice were pretreated with compound 8. The 30-day mortality assay, hematoxylin-eosin staining, and immunohistochemistry staining assay were performed to evaluate the anti-radiation effect of the compound 8. TUNEL and immunofluorescence assays were conducted to study the anti-radiation mechanism of compound 8. RESULTS Compared to the IR + vehicle group, the 30-day survival rate of mice treated with 25 mg/kg of compound 8 was significantly improved after 8 Gy total body irradiation. In the morphological study of the small intestine, we found that compound 8 could maintain crypt-villus structures in the irradiated mice. Further immunohistochemical staining displayed that compound 8 could improve the survival of Lgr5+ cells, ki67+ cells, and lysozyme+ cells. The results of TUNEL and immunofluorescence assays showed that compound 8 could decrease the expression of apoptosis-related caspase-8/-9, γ-H2AX, Bax, and p53. CONCLUSIONS These results indicate that compound 8 exerts its effects by maintaining structure and function of small intestine. It also reduces DNA damage, promotes crypt proliferation and differentiation. Moreover, it may enhance the anti-apoptotic ability of small intestinal tissue by inhibiting the activation of p53 and blocking the caspase cascade reaction. Compound 8 can protect the intestinal tract from post-radiation damage, it is thus a new and effective protective agent of radiation.
Collapse
Affiliation(s)
- Xinxin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Renbin Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Longfei Miao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Xuejiao Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| | - Hongqi Tian
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, China
| |
Collapse
|
5
|
Bachman JF, Chakkalakal JV. Insights into muscle stem cell dynamics during postnatal development. FEBS J 2022; 289:2710-2722. [PMID: 33811430 PMCID: PMC9947813 DOI: 10.1111/febs.15856] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/17/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022]
Abstract
During development, resident stem cell populations contribute to the growth and maturation of tissue and organs. In skeletal muscle, muscle stem cells, or satellite cells (SCs), are responsible for the maturation of postnatal myofibers. However, the role SCs play in later stages of postnatal growth, and thus, when they enter a mature quiescent state is controversial. Here, we discuss the current literature regarding the role SCs play in all stages of postnatal growth, from birth to puberty onset to young adulthood. We additionally highlight the implications of SC loss or dysfunction during developmental stages, both in the context of experimental paradigms and disease settings.
Collapse
Affiliation(s)
- John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester NY, United States.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester NY, United States
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester NY, United States.,Department of Biomedical Engineering, University of Rochester, Rochester NY, United States.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester NY, United States.,Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester NY, United States.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester NY, United States
| |
Collapse
|
6
|
Kallenbach JG, Bachman JF, Paris ND, Blanc RS, O'Connor T, Furati E, Williams JP, Chakkalakal JV. Muscle-specific functional deficits and lifelong fibrosis in response to paediatric radiotherapy and tumour elimination. J Cachexia Sarcopenia Muscle 2022; 13:296-310. [PMID: 34997696 PMCID: PMC8818600 DOI: 10.1002/jcsm.12902] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/28/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND As paediatric cancer survivors are living into adulthood, they suffer from the age-related, accelerated decline of functional skeletal muscle tissue, termed sarcopenia. With ionizing radiation (radiotherapy) at the core of paediatric cancer therapies, its direct and indirect effects can have lifelong negative impacts on paediatric growth and maintenance of skeletal muscle. Utilizing our recently developed preclinical rhabdomyosarcoma mouse model, we investigated the late effects of paediatric radiation treatment on skeletal muscles from late adolescent (8 weeks old) and middle-aged (16 months old) mice. METHODS Paediatric C57BL/6J male mice (3 weeks old) were injected with rhabdomyosarcoma cells into their right hindlimbs, and then fractionated irradiation (3 × 8.2 Gy) was administered to those limbs at 4 weeks old to eliminate the tumours. Radiation-alone and tumour-irradiated mice were assessed at either 8 weeks (3 weeks post-irradiation) or 16 months (14 months post-irradiation) of age for muscle physiology, myofibre characteristics, cell loss, histopathology, fibrosis, inflammatory gene expression, and fibrotic gene expression. RESULTS Mice that received only paediatric radiation demonstrated reduced muscle mass (-17%, P < 0.001), muscle physiological function (-25%, P < 0.01), muscle contractile kinetics (-25%, P < 0.05), satellite cell number (-45%, P < 0.05), myofibre cross-sectional area (-30%, P < 0.0001), and myonuclear number (-17%, P < 0.001). Paediatric radiation increased inflammatory gene expression, increased fibrotic gene expression, and induced extracellular matrix protein deposition (fibrosis) with tumour elimination exacerbating some phenotypes. Paediatric tumour-eliminated mice demonstrated exacerbated deficits to function (-20%, P < 0.05) and myofibre size (-17%, P < 0.001) in some muscles as well as further increases to inflammatory and fibrotic gene expression. Examining the age-related effects of paediatric radiotherapy in middle-aged mice, we found persistent myofibre atrophy (-20%, P < 0.01), myonuclear loss (-18%, P < 0.001), up-regulated inflammatory and fibrotic signalling, and lifelong fibrosis. CONCLUSIONS The results from this paediatric radiotherapy model are consistent and recapitulate the clinical and molecular features of accelerated sarcopenia, musculoskeletal frailty, and radiation-induced fibrosis experienced by paediatric cancer survivors. We believe that this preclinical mouse model is well poised for future mechanistic insights and therapeutic interventions that improve the quality of life for paediatric cancer survivors.
Collapse
Affiliation(s)
- Jacob G Kallenbach
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas O'Connor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Esraa Furati
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacqueline P Williams
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.,Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Joe V Chakkalakal
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopedic Surgery and Cell Biology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
7
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
8
|
Singh VK, Seed TM. Repurposing Pharmaceuticals Previously Approved by Regulatory Agencies to Medically Counter Injuries Arising Either Early or Late Following Radiation Exposure. Front Pharmacol 2021; 12:624844. [PMID: 34040517 PMCID: PMC8141805 DOI: 10.3389/fphar.2021.624844] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
The increasing risks of radiological or nuclear attacks or associated accidents have served to renew interest in developing radiation medical countermeasures. The development of prospective countermeasures and the subsequent gain of Food and Drug Administration (FDA) approval are invariably time consuming and expensive processes, especially in terms of generating essential human data. Due to the limited resources for drug development and the need for expedited drug approval, drug developers have turned, in part, to the strategy of repurposing agents for which safety and clinical data are already available. Approval of drugs that are already in clinical use for one indication and are being repurposed for another indication is inherently faster and more cost effective than for new agents that lack regulatory approval of any sort. There are four known growth factors which have been repurposed in the recent past as radiomitigators following the FDA Animal Rule: Neupogen, Neulasta, Leukine, and Nplate. These four drugs were in clinic for several decades for other indications and were repurposed. A large number of additional agents approved by various regulatory authorities for given indications are currently under investigation for dual use for acute radiation syndrome or for delayed pathological effects of acute radiation exposure. The process of drug repurposing, however, is not without its own set of challenges and limitations.
Collapse
Affiliation(s)
- Vijay K. Singh
- Division of Radioprotectants, Department of Pharmacology and Molecular Therapeutics, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | | |
Collapse
|
9
|
Chancellor J, Nowadly C, Williams J, Aunon-Chancellor S, Chesal M, Looper J, Newhauser W. Everything you wanted to know about space radiation but were afraid to ask. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:113-128. [PMID: 33902392 DOI: 10.1080/26896583.2021.1897273] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The space radiation environment is a complex combination of fast-moving ions derived from all atomic species found in the periodic table. The energy spectrum of each ion species varies widely but is prominently in the range of 400-600 MeV/n. The large dynamic range in ion energy is difficult to simulate in ground-based radiobiology experiments. Most ground-based irradiations with mono-energetic beams of a single one ion species are delivered at comparatively high dose rates. In some cases, sequences of such beams are delivered with various ion species and energies to crudely approximate the complex space radiation environment. This approximation may cause profound experimental bias in processes such as biologic repair of radiation damage, which are known to have strong temporal dependencies. It is possible that this experimental bias leads to an over-prediction of risks of radiation effects that have not been observed in the astronaut cohort. None of the primary health risks presumably attributed to space radiation exposure, such as radiation carcinogenesis, cardiovascular disease, cognitive deficits, etc., have been observed in astronaut or cosmonaut crews. This fundamentally and profoundly limits our understanding of the effects of GCR on humans and limits the development of effective radiation countermeasures.
Collapse
Affiliation(s)
- Jeffery Chancellor
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Preventive Medicine & Population Health, University of Texas Medical Branch, Galveston, Texas, USA
- Outer Space Insititute, Universit of British Columbia, CA
| | - Craig Nowadly
- Department of Emergency Medicine, Brooke Army Medical Center, Fort Sam Houston, Texas, USA
| | - Jacqueline Williams
- Departments of Environmental Medicine & Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - Serena Aunon-Chancellor
- Department of Preventive Medicine & Population Health, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Internal Medicine, LSU Health Science Center, Baton Rouge, Louisiana, USA
- Astronaut Office, NASA Johnson Space Center, Houston, Texas, USA
| | - Megan Chesal
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Jayme Looper
- Department of Veterinary Clinical Sciences, LSU School of Veterinary Medicine, Baton Rouge, Louisiana, USA
| | - Wayne Newhauser
- Department of Physics & Astronomy, Louisiana State University, Baton Rouge, Louisiana, USA
- Department of Physics, Mary Bird Perkins Cancer Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
10
|
Chemoradiation impairs myofiber hypertrophic growth in a pediatric tumor model. Sci Rep 2020; 10:19501. [PMID: 33177579 PMCID: PMC7659015 DOI: 10.1038/s41598-020-75913-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
Pediatric cancer treatment often involves chemotherapy and radiation, where off-target effects can include skeletal muscle decline. The effect of such treatments on juvenile skeletal muscle growth has yet to be investigated. We employed a small animal irradiator to administer fractionated hindlimb irradiation to juvenile mice bearing implanted rhabdomyosarcoma (RMS) tumors. Hindlimb-targeted irradiation (3 × 8.2 Gy) of 4-week-old mice successfully eliminated RMS tumors implanted one week prior. After establishment of this preclinical model, a cohort of tumor-bearing mice were injected with the chemotherapeutic drug, vincristine, alone or in combination with fractionated irradiation (5 × 4.8 Gy). Single myofiber analysis of fast-contracting extensor digitorum longus (EDL) and slow-contracting soleus (SOL) muscles was conducted 3 weeks post-treatment. Although a reduction in myofiber size was apparent, EDL and SOL myonuclear number were differentially affected by juvenile irradiation and/or vincristine treatment. In contrast, a decrease in myonuclear domain (myofiber volume/myonucleus) was observed regardless of muscle or treatment. Thus, inhibition of myofiber hypertrophic growth is a consistent feature of pediatric cancer treatment.
Collapse
|
11
|
Bachman JF, Blanc RS, Paris ND, Kallenbach JG, Johnston CJ, Hernady E, Williams JP, Chakkalakal JV. Radiation-Induced Damage to Prepubertal Pax7+ Skeletal Muscle Stem Cells Drives Lifelong Deficits in Myofiber Size and Nuclear Number. iScience 2020; 23:101760. [PMID: 33241204 PMCID: PMC7674517 DOI: 10.1016/j.isci.2020.101760] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/08/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
During prepubertal development, muscle stem cells (satellite cells, SCs) actively contribute to myofiber growth. Because some SCs are active during this time, they may be particularly susceptible to damage. Using a Small Animal Radiation Research Platform (SARRP), we investigated the effects of local fractionated radiation treatment on prepubertal SCs. Immediately after this regimen, there was a reduction in SC number. Although surviving SCs had deficiencies in function, some myogenic potential remained. Indeed, some muscle regenerative capacity persisted immediately after irradiation. Lastly, we assessed the long-term consequences of radiation-induced SC loss during prepuberty. We observed a reduction of myofiber size and corresponding loss of nuclei in both fast- and slow-contracting muscles 14 months post-irradiation. Notably, prepubertal SC depletion mimicked these lifelong deficits. This work highlights the susceptibility of prepubertal SCs to radiation exposure. We also reveal the importance of prepubertal SC contribution to the lifelong maintenance of skeletal muscle. Increased sensitivity of satellite cells to irradiation during prepubertal growth Prepubertal irradiation leads to lifelong deficits in skeletal muscle regenerative capacity Lifelong reduction in myofiber size and nuclear number is a consequence of prepubertal irradiation Satellite cell ablation mimics the lifelong effects of prepubertal irradiation on myofiber size and nuclear number
Collapse
Affiliation(s)
- John F Bachman
- Department of Pathology and Laboratory Medicine, Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Roméo S Blanc
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nicole D Paris
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacob G Kallenbach
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Carl J Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Eric Hernady
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacqueline P Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Joe V Chakkalakal
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA.,Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.,Stem Cell and Regenerative Medicine Institute, and The Rochester Aging Research Center, University of Rochester Medical Center, Rochester, NY, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
12
|
Daniel AR, Lee CL, Oh P, Luo L, Ma Y, Kirsch DG. Inhibiting Glycogen Synthase Kinase-3 Mitigates the Hematopoietic Acute Radiation Syndrome in a Sex- and Strain-dependent Manner in Mice. HEALTH PHYSICS 2020; 119:315-321. [PMID: 32175929 PMCID: PMC7398824 DOI: 10.1097/hp.0000000000001243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The Radiation and Nuclear Countermeasures Program at the National Institute of Allergy and Infectious Diseases (NIAID) mandated that medical countermeasures for treating Acute Radiation Syndrome (ARS) must have efficacy when administered at least 24 h after radiation exposure. At this time point, many cells within key target tissues, such as the hematopoietic system and the gastrointestinal (GI) tract, will already be dead. Therefore, drugs that promote the regeneration of surviving cells may improve outcomes. The serine/threonine kinase glycogen synthase kinase-3 (GSK-3) regulates stem and progenitor cell self-renewal and regeneration in the hematopoietic and GI compartments. We tested inhibition of GSK-3β by SB216763 24 h after total body irradiation (TBI) and sub-total body irradiation (SBI). Here, we show that subcutaneous administration of SB216763 promotes the regeneration of surviving hematopoietic stem/progenitor cells (HSPCs), including myeloid progenitor cells, and improves survival of C57Bl/6 male mice when administered 24 h after TBI. However, these results were not recapitulated in female C57Bl/6 animals, suggesting a sex difference in GSK-3β signaling in HSPCs. Subcutaneous administration of SB216763 in male mice stimulated activation of Sox2 transcription but failed to induce Sox2 transcription in female C57Bl/6 mice. Using TCF/lef-GFP reporter mice, we examined Wnt signaling in HSPCs of irradiated male and female mice treated with SB216763. GSK-3 inhibition elevated Wnt reporter activity in HSPCs isolated from male but not female mice. SB216763 did not mitigate hematopoietic ARS in males or females of a second strain of wild-type mice, C3H. In addition, administration of SB216763 did not mitigate hematopoietic ARS beyond the currently available standard approved therapy of ciprofloxacin and granulocyte-colony stimulating factor (G-CSF) in male C57Bl/6 mice. Further, SB216763 did not mitigate GI-ARS after SBI in C57Bl/6 male mice. The lack of efficacy in both sexes and multiple strains of mice indicate that SB216763 is not suitable for further drug development as a mitigator of ARS. Our studies demonstrate that activation of Wnt signaling in HSPCs promotes hematopoietic regeneration following radiation exposure, and targeting this pathway downstream of GSK-3β may mitigate ARS in a sex- and strain-independent manner.
Collapse
Affiliation(s)
- Andrea R. Daniel
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Patrick Oh
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - David G. Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina 27710, USA
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
13
|
Cranial irradiation acutely and persistently impairs injury-induced microglial proliferation. Brain Behav Immun Health 2020; 4:100057. [PMID: 34589843 PMCID: PMC8474291 DOI: 10.1016/j.bbih.2020.100057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/28/2020] [Accepted: 03/01/2020] [Indexed: 12/12/2022] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), play multiple roles in maintaining CNS homeostasis and mediating tissue repair, including proliferating in response to brain injury and disease. Cranial irradiation (CI), used for the treatment of brain tumors, has a long-lasting anti-proliferative effect on a number of cell types in the brain, including oligodendrocyte progenitor and neural progenitor cells; however, the effect of CI on CNS-resident microglial proliferation is not well characterized. Using a sterile cortical needle stab injury model in mice, we found that the ability of CNS-resident microglia to proliferate in response to injury was impaired by prior CI, in a dose-dependent manner, and was nearly abolished by a 20 Gy dose. Similarly, in a metastatic tumor model, prior CI (20 Gy) reduced microglial proliferation in response to tumor growth. The effect of irradiation was long-lasting; 20 Gy CI 6 months prior to stab injury significantly impaired microglial proliferation. We also investigated how stab and/or irradiation impacted levels of P2Y12R, CD68, CSF1, IL-34 and CSF1R, factors involved in the brain’s normal response to injury. P2Y12R, CD68, CSF1, and IL-34 expression were altered by stab similarly in irradiated mice and controls; however, CSF1R was differentially affected. qRT-PCR and flow cytometry analyses demonstrated that CI reduced overall Csf1r mRNA levels and microglial specific CSF1R protein expression, respectively. Interestingly, Csf1r mRNA levels increased after injury in unirradiated controls; however, Csf1r levels were persistently decreased in irradiated mice, and did not increase in response to stab. Together, our data demonstrate that CI leads to a significant and lasting impairment of microglial proliferation, possibly through a CSF1R-mediated mechanism. Irradiation leads to a long-term deficit in injury-induced microglial proliferation. Irradiation reduces microglial proliferation associated with tumor growth. Irradiation decreases microglial CSF1R and prevents its upregulation after injury.
Collapse
|
14
|
Camara Planek MI, Silver AJ, Volgman AS, Okwuosa TM. Exploratory Review of the Role of Statins, Colchicine, and Aspirin for the Prevention of Radiation-Associated Cardiovascular Disease and Mortality. J Am Heart Assoc 2020; 9:e014668. [PMID: 31960749 PMCID: PMC7033839 DOI: 10.1161/jaha.119.014668] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Adam J. Silver
- Rush Heart Center for WomenRush University Medical CenterChicagoIL
| | | | | |
Collapse
|
15
|
Beach TA, Groves AM, Williams JP, Finkelstein JN. Modeling radiation-induced lung injury: lessons learned from whole thorax irradiation. Int J Radiat Biol 2020; 96:129-144. [PMID: 30359147 PMCID: PMC6483900 DOI: 10.1080/09553002.2018.1532619] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/25/2022]
Abstract
Models of thoracic irradiation have been developed as clinicians and scientists have attempted to decipher the events that led up to the pulmonary toxicity seen in human subjects following radiation treatment. The most common model is that of whole thorax irradiation (WTI), applied in a single dose. Mice, particularly the C57BL/6J strain, has been frequently used in these investigations, and has greatly informed our current understanding of the initiation and progression of radiation-induced lung injury (RILI). In this review, we highlight the sequential progression and dynamic nature of RILI, focusing primarily on the vast array of information that has been gleaned from the murine model. Ample evidence indicates a wide array of biological responses that can be seen following irradiation, including DNA damage, oxidative stress, cellular senescence and inflammation, all triggered by the initial exposure to ionizing radiation (IR) and heterogeneously maintained throughout the temporal progression of injury, which manifests as acute pneumonitis and later fibrosis. It appears that the early responses of specific cell types may promote further injury, disrupting the microenvironment and preventing a return to homeostasis, although the exact mechanisms driving these responses remains somewhat unclear. Attempts to either prevent or treat RILI in preclinical models have shown some success by targeting these disparate radiobiological processes. As our understanding of the dynamic cellular responses to radiation improves through the use of such models, so does the likelihood of preventing or treating RILI.
Collapse
Affiliation(s)
- Tyler A. Beach
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Angela M. Groves
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
16
|
Emmons R, Ngu M, Xu G, Hernández-Saavedra D, Chen H, DE Lisio M. Effects of Obesity and Exercise on Bone Marrow Progenitor Cells after Radiation. Med Sci Sports Exerc 2019; 51:1126-1136. [PMID: 30640286 DOI: 10.1249/mss.0000000000001894] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION The late effects of radiation therapy can have significant consequences for the health and quality of life of long-term cancer survivors. Radiation induces persistent alterations in hematopoietic stem and progenitor cells (HSPC) and the bone marrow environment; however, how relevant host factors such as obesity and exercise differentially regulate HSPC content and the bone marrow environment after radiation exposure remains unknown. The purpose of this investigation was to evaluate how the combination of obesity and exercise training modulates HSPC and their niche after sublethal radiation exposure in mice. METHODS Mice fed either a control or a high-fat diet to induce obesity remained sedentary or underwent a progressive treadmill exercise program. At 13 wk of age, mice were irradiated (3 Gy) and continued their specific diets and exercise program for four more weeks. RESULTS Exercise-trained mice had significantly higher quantities of several HSPC subpopulations and bone marrow stromal cell populations, whereas HSPC subpopulations were significantly lower in obese mice after radiation. Reactive oxygen species content was significantly decreased in HSPC with exercise training. Proteomics analysis of bone marrow supernatant revealed clustering of biologically relevant changes in exercise-trained mice. Functional evaluation of bone marrow supernatant revealed a significant increase in leukemia blast viability in obese mice but not in the exercise-trained mice (P < 0.05). CONCLUSION Together, these data suggest that exercise training partially restores the negative effects of obesity on HSPC and their niche after radiation exposure. As such, exercise training should be considered to mitigate the late effects of radiation therapy on the hematopoietic system for cancer survivors with or without obesity who have undergone radiation therapy.
Collapse
Affiliation(s)
- Russell Emmons
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Matthew Ngu
- School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA
| | - Guanying Xu
- Department of Food Sciences and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL
| | | | - Hong Chen
- Department of Food Sciences and Human Nutrition, University of Illinois at Urbana-Champaign, Champaign, IL.,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL
| | - Michael DE Lisio
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Champaign, IL.,School of Human Kinetics, University of Ottawa, Ottawa, ON, CANADA.,Regenerative Medicine Program, Centre on Neuromuscular Disease, and Brain and Mind Institute, University of Ottawa, Ottawa, ON, CANADA
| |
Collapse
|
17
|
C/EBPδ protects from radiation-induced intestinal injury and sepsis by suppression of inflammatory and nitrosative stress. Sci Rep 2019; 9:13953. [PMID: 31562350 PMCID: PMC6764943 DOI: 10.1038/s41598-019-49437-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Abstract
Ionizing radiation (IR)-induced intestinal damage is characterized by a loss of intestinal crypt cells, intestinal barrier disruption and translocation of intestinal microflora resulting in sepsis-mediated lethality. We have shown that mice lacking C/EBPδ display IR-induced intestinal and hematopoietic injury and lethality. The purpose of this study was to investigate whether increased IR-induced inflammatory, oxidative and nitrosative stress promote intestinal injury and sepsis-mediated lethality in Cebpd−/− mice. We found that irradiated Cebpd−/− mice show decreased villous height, crypt depth, crypt to villi ratio and expression of the proliferation marker, proliferating cell nuclear antigen, indicative of intestinal injury. Cebpd−/− mice show increased expression of the pro-inflammatory cytokines (Il-6, Tnf-α) and chemokines (Cxcl1, Mcp-1, Mif-1α) and Nos2 in the intestinal tissues compared to Cebpd+/+ mice after exposure to TBI. Cebpd−/− mice show decreased GSH/GSSG ratio, increased S-nitrosoglutathione and 3-nitrotyrosine in the intestine indicative of basal oxidative and nitrosative stress, which was exacerbated by IR. Irradiated Cebpd-deficient mice showed upregulation of Claudin-2 that correlated with increased intestinal permeability, presence of plasma endotoxin and bacterial translocation to the liver. Overall these results uncover a novel role for C/EBPδ in protection against IR-induced intestinal injury by suppressing inflammation and nitrosative stress and underlying sepsis-induced lethality.
Collapse
|
18
|
Calvi LM, Frisch BJ, Kingsley PD, Koniski AD, Love TM, Williams JP, Palis J. Acute and late effects of combined internal and external radiation exposures on the hematopoietic system. Int J Radiat Biol 2019; 95:1447-1461. [PMID: 31329495 DOI: 10.1080/09553002.2019.1644932] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Purpose: Incidents, such as nuclear facility accidents and the release of a 'dirty bomb', might result in not only external irradiation of personnel, but additional internal exposures through concomitant inhalation and/or ingestion of radioactive particulates. The purpose of this study was to define the impact of such a combination of radiation injuries on the hematopoietic niche.Material and methods: To assess changes in the murine hematopoietic system, we used a combined exposure of total body irradiation (TBI, 6 Gy) followed immediately by an internal (intraperitoneal) administration of 100 µCi of soluble 137Cs. We then evaluated acute survival in combined versus single modality exposure groups, as well as assessing hematopoietic function at 12 and 26 week time points.Results: Acutely, the combination of external and internal exposures led to an unexpected delay in excretion of 137Cs, increasing the absorbed dose in the combined exposure group and leading to mortality from an acute hematopoietic syndrome. At 12 weeks, all exposure paradigms resulted in decreased numbers of phenotypic hematopoietic stem cells (HSCs), particularly the short-term HSCs (ST-HSC); long-term HSCs (LT-HSC) were depleted only in the internal and combined exposure groups. At 26 weeks, there was significant anemia in both the TBI alone and combined exposure groups. There were decreased numbers in both the LT- and ST-HSCs and decreased functionality, as measured by competitive repopulation, was seen in all radiation groups, with the greatest effects seen in the internal and combined exposure groups.Conclusions: Our data indicate that a combined injury of sublethal external irradiation with internal contamination induces significant and persistent changes in the hematopoietic system, as may have been predicted from the literature and our own group's findings. However, a novel observation was that the combined exposure led to an alteration in the excretion kinetics of the internal contamination, increasing the acute effects beyond those anticipated. As a result, we believe that a combined exposure poses a unique challenge to the medical community during both the acute and, possibly, delayed recovery stages.
Collapse
Affiliation(s)
- Laura M Calvi
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Benjamin J Frisch
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Paul D Kingsley
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Anne D Koniski
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| | - Tanzy M Love
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
| | - Jacqueline P Williams
- Department of Environmental Medicine and Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - James Palis
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
19
|
Groves AM, Williams JP. Saving normal tissues - a goal for the ages. Int J Radiat Biol 2019; 95:920-935. [PMID: 30822213 PMCID: PMC7183326 DOI: 10.1080/09553002.2019.1589654] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 02/08/2023]
Abstract
Almost since the earliest utilization of ionizing radiation, many within the radiation community have worked toward either preventing (i.e. protecting) normal tissues from unwanted radiation injury or rescuing them from the downstream consequences of exposure. However, despite over a century of such investigations, only incremental gains have been made toward this goal and, with certainty, no outright panacea having been found. In celebration of the 60th anniversary of the International Journal of Radiation Biology and to chronicle the efforts that have been made to date, we undertook a non-rigorous survey of the articles published by normal tissue researchers in this area, using those that have appeared in the aforementioned journal as a road map. Three 'snapshots' of publications on normal tissue countermeasures were taken: the earliest (1959-1963) and most recent (2013-2018) 5-year of issues, as well as a 5-year intermediate span (1987-1991). Limiting the survey solely to articles appearing within International Journal of Radiation Biology likely reduced the number of translational studies interrogated given the basic science tenor of this particular publication. In addition, by taking 'snapshots' rather than considering the entire breadth of the journal's history in this field, important papers that were published during the interim periods were omitted, for which we apologize. Nonetheless, since the journal's inception, we observed that, during the chosen periods, the majority of studies undertaken in the field of normal tissue countermeasures, whether investigating radiation protectants, mitigators or treatments, have focused on agents that interfere with the physical, chemical and/or biological effects known to occur during the acute period following whole body/high single dose exposures. This relatively narrow approach to the reduction of normal tissue effects, especially those that can take months, if not years, to develop, seems to contradict our growing understanding of the progressive complexities of the microenvironmental disruption that follows the initial radiation injury. Given the analytical tools now at our disposal and the enormous benefits that may be reaped in terms of improving patient outcomes, as well as the potential for offering countermeasures to those affected by accidental or mass casualty exposures, it appears time to broaden our approaches to developing normal tissue countermeasures. We have no doubt that the contributors and readership of the International Journal of Radiation Biology will continue to contribute to this effort for the foreseeable future.
Collapse
Affiliation(s)
- Angela M. Groves
- Departments of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, USA
| | - Jacqueline P. Williams
- Departments of Environmental Medicine, University of Rochester Medical Center, Rochester, USA
- Departments of Radiation Oncology, University of Rochester Medical Center, Rochester, USA
| |
Collapse
|
20
|
Beach TA, Groves AM, Johnston CJ, Williams JP, Finkelstein JN. Recurrent DNA damage is associated with persistent injury in progressive radiation-induced pulmonary fibrosis. Int J Radiat Biol 2018; 94:1104-1115. [PMID: 30238842 PMCID: PMC6309234 DOI: 10.1080/09553002.2018.1516907] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/17/2018] [Accepted: 08/21/2018] [Indexed: 12/19/2022]
Abstract
PURPOSE Radiation-induced lung injuries (RILI), namely radiation pneumonitis and/or fibrosis, are dose-limiting outcomes following treatment for thoracic cancers. As part of a search for mitigation targets, we sought to determine if persistent DNA damage is a characteristic of this progressive injury. METHODS C57BL/6J female mice were sacrificed at 24 h, 1, 4, 12, 16, 24 and 32 weeks following a single dose of 12.5 Gy thorax only gamma radiation; their lungs were compared to age-matched unirradiated animals. Tissues were examined for DNA double-strand breaks (DSBs) (γ-H2A.X and p53bp1), cellular senescence (senescence-associated beta-galactosidase and p21) and oxidative stress (malondialdehyde). RESULTS Data revealed consistently higher numbers of DSBs compared to age-matched controls, with increases in γ-H2A.X positivity beyond 24 h post-exposure, particularly during the pathological phases, suggesting periods of recurrent DNA damage. Additional intermittent increases in both cellular senescence and oxidative stress also appeared to coincide with pneumonitis and fibrosis. CONCLUSIONS These novel, long-term data indicate (a) increased and persistent levels of DSBs, oxidative stress and cellular senescence may serve as bioindicators of RILI, and (b) prevention of genotoxicity, via mitigation of free radical production, continues to be a potential strategy for the prevention of pulmonary radiation injury.
Collapse
Affiliation(s)
- Tyler A. Beach
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
| | - Angela M. Groves
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Carl J. Johnston
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
21
|
Groves AM, Williams JP, Hernady E, Reed C, Fenton B, Love T, Finkelstein JN, Johnston CJ. A Potential Biomarker for Predicting the Risk of Radiation-Induced Fibrosis in the Lung. Radiat Res 2018; 190:513-525. [PMID: 30117783 DOI: 10.1667/rr15122.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Biomarkers could play an essential role during triage in the aftermath of a radiological event, where exposure to radiation will be heterogeneous and complicated by concurrent trauma. Used alongside biodosimetry, biomarkers can identify victims in need of treatment for acute radiation effects, and might also provide valuable information on later developing consequences that need to be addressed as part of a treatment strategy. Indeed, because the lung is particularly sensitive to radiation and resultant late effects not only affect quality of life, but can also lead to morbidity, the risk of developing downstream pulmonary complications in exposed individuals requires assessment. In this study, analyses of changes in pulmonary and circulating content of club cell secretory protein (CCSP) and surfactant protein D (SP-D), expressed by epithelial club cells and type II pneumocytes in the lung, respectively, were used to evaluate pulmonary epithelial damage in several lung injury models. Using a combined radiation exposure model, fibrosis-susceptible C57BL/6J (C57) and alveolitis-prone C3H/HeJ (C3H) mice received 5 Gy total-body irradiation plus 2.5-10 Gy whole-lung irradiation, and lung and plasma samples were collected throughout the course of the radiation response, at time points ranging from 24 h to 26 weeks postirradiation. Radiation significantly reduced bronchiole CCSP coverage in C57 mice at 26 weeks, a response that varied in extent among animals, but correlated with the severity of fibrosis in each animal. Interestingly, plasma CCSP content was elevated in C57 mice at multiple time points preceding and during the fibrotic period; this response that was not observed in C3H mice. Circulating CCSP/SP-D ratios, calculated as an index of lung integrity, were similarly increased throughout the time course in C57, but not C3H, mice. Furthermore, when the thoracic doses were reduced to subthreshold levels for fibrosis induction (2.5 or 7.5 Gy), although the CCSP/SP-D ratio in lung homogenates demonstrated dose-responsive changes, this was not reflected in the plasma ratios at acute and late time points. Importantly, plasma CCSP/SP-D ratios also were not significantly altered in C57 mice exposed to LPS, and only transiently decreased in influenza-exposed mice, demonstrating a level of specificity for radiation-induced lung injury. These results indicate that the CCSP/SP-D ratio, measured in plasma, is sensitive to individual variation in radiation sensitivity, correlates with fibrosis development, can be detected early after exposure and is specific to radiation-induced injury. This suggests that the CCSP/SP-D ratio may be useful as a biomarker of radiation-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Angela M Groves
- Departments of a Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York
| | - Jacqueline P Williams
- b Environmental Medicine, University of Rochester Medical Center, Rochester, New York.,c Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| | - Eric Hernady
- b Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Christina Reed
- Departments of a Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York
| | - Bruce Fenton
- c Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| | - Tanzy Love
- d Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York
| | - Jacob N Finkelstein
- Departments of a Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York.,b Environmental Medicine, University of Rochester Medical Center, Rochester, New York.,c Radiation Oncology, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Departments of a Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York.,b Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
22
|
Williams JP, Newhauser W. Normal tissue damage: its importance, history and challenges for the future. Br J Radiol 2018; 92:20180048. [PMID: 29616836 DOI: 10.1259/bjr.20180048] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Sir Oliver Scott, a philanthropist and radiation biologist and, therefore, the epitome of a gentleman and a scholar, was an early Director of the BECC Radiobiology Research Unit at Mount Vernon. His tenure preceded that of Jack Fowler, with both contributing to basic, translational and clinical thought and application in radiation across the globe. With respect to this review, Fowler's name in particular has remained synonymous with the use of models, both animal and mathematical, that assess and quantify the biological mechanisms that underlie radiation-associated normal tissue toxicities. An understanding of these effects is critical to the optimal use of radiation therapy in the clinic; however, the role that basic sciences play in clinical practice has been undergoing considerable change in recent years, particularly in the USA, where there has been a growing emphasis on engineering and imaging to improve radiation delivery, with empirical observations of clinical outcome taking the place of models underpinned by evidence from basic science experiments. In honour of Scott and Fowler's work, we have taken this opportunity to review how our respective fields of radiation biology and radiation physics have intertwined over the years, affecting the clinical use of radiation with respect to normal tissue outcomes. We discuss the past and current achievements, with the hope of encouraging a revived interest in physics and biology as they relate to radiation oncology practice, since, like Scott and Fowler, we share the goal of improving the future outlook for cancer patients.
Collapse
Affiliation(s)
- Jacqueline P Williams
- Departments of Environmental Medicine and Radiation Oncology, University of Rochester Medical Center, Rochester, NY, USA
| | - Wayne Newhauser
- Department of Physics and Astronomy, Louisiana State University, Baton Rouge, LA, USA
| |
Collapse
|
23
|
Groves AM, Johnston CJ, Williams JP, Finkelstein JN. Role of Infiltrating Monocytes in the Development of Radiation-Induced Pulmonary Fibrosis. Radiat Res 2018; 189:300-311. [PMID: 29332538 DOI: 10.1667/rr14874.1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Lung exposure to radiation induces an injury response that includes the release of cytokines and chemotactic mediators; these signals recruit immune cells to execute inflammatory and wound-healing processes. However, radiation alters the pulmonary microenvironment, dysregulating the immune responses and preventing a return to homeostasis. Importantly, dysregulation is observed as a chronic inflammation, which can progress into pneumonitis and promote pulmonary fibrosis; inflammatory monocytes, which are bone marrow derived and express CCR2, have been shown to migrate into the lung after radiation exposure. Although the extent to which recruited inflammatory monocytes contribute to radiation-induced pulmonary fibrosis has not been fully investigated, we hypothesize that its pathogenesis is reliant on this population. The CC chemokine ligand, CCL2, is a chemotactic mediator responsible for trafficking of CCR2+ inflammatory cells into the lung. Therefore, the contribution of this mediator to fibrosis development was analyzed. Interleukin (IL)-1β, a potent pro-inflammatory cytokine expressed during the radiation response, and its receptor, IL-1R1, were also evaluated. To this end, CCR2-/-, IL-1β-/- and IL-1R1-/- chimeric mice were generated and exposed to 12.5 Gy thoracic radiation, and their response was compared to wild-type (C57BL/6) syngeneic controls. Fibrotic foci were observed in the periphery of the lungs of C57 syngeneic mice and CCR2-/- recipient mice that received C57 bone marrow (C57 > CCR2-/-) by 16 and 12 weeks after irradiation, respectively. In contrast, in the mice that had received bone marrow lacking CCR2 (CCR2-/- > C57 and CCR2-/- syngeneic mice), no pulmonary fibrosis was observed at 22 weeks postirradiation. This observation correlated with decreased numbers of infiltrating and interstitial macrophages compared to controls, as well as reduced proportions of pro-inflammatory Ly6C+ macrophages observed at 12-18 weeks postirradiation, suggesting that CCR2+ macrophages contribute to radiation-induced pulmonary fibrosis. Interestingly, reduced proportions of CD206+ lung macrophages were also present at these time points in CCR2-/- chimeric mice, regardless of donor bone marrow type, suggesting that the phenotype of resident subsets may be influenced by CCR2. Furthermore, chimeras, in which either IL-1β was ablated from infiltrating cells or IL-1R1 from lung tissues, were also protected from fibrosis development, correlating with attenuated CCL2 production; these data suggest that IL-1β may influence chemotactic signaling after irradiation. Overall, our data suggest that CCR2+ infiltrating monocyte-derived macrophages may play a critical role in the development of radiation-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Angela M Groves
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York
| | - Carl J Johnston
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jacob N Finkelstein
- Department of a Pediatrics M&D Neonatology, University of Rochester Medical Center, Rochester, New York.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
24
|
McLaughlin MF, Donoviel DB, Jones JA. Novel Indications for Commonly Used Medications as Radiation Protectants in Spaceflight. Aerosp Med Hum Perform 2017. [PMID: 28641684 DOI: 10.3357/amhp.4735.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND In the space environment, the traditional radioprotective principles of time, distance, and shielding become difficult to implement. Additionally, the complex radiation environment inherent in space, the chronic exposure timeframe, and the presence of numerous confounding variables complicate the process of creating appropriate risk models for astronaut exposure. Pharmaceutical options hold tremendous promise to attenuate acute and late effects of radiation exposure in the astronaut population. Pharmaceuticals currently approved for other indications may also offer radiation protection, modulation, or mitigation properties along with a well-established safety profile. Currently there are only three agents which have been clinically approved to be employed for radiation exposure, and these only for very narrow indications. This review identifies a number of agents currently approved by the U.S. Food and Drug Administration (FDA) which could warrant further investigation for use in astronauts. Specifically, we examine preclinical and clinical evidence for statins, nonsteroidal anti-inflammatory drugs (NSAIDs), angiotensin converting enzyme inhibitors (ACEIs), angiotensin II receptor blockers (ARBs), metformin, calcium channel blockers, β adrenergic receptor blockers, fingolimod, N-acetylcysteine, and pentoxifylline as potential radiation countermeasures.McLaughlin MF, Donoviel DB, Jones JA. Novel indications for commonly used medications as radiation protectants in spaceflight. Aerosp Med Hum Perform. 2017; 88(7):665-676.
Collapse
|
25
|
Beach TA, Johnston CJ, Groves AM, Williams JP, Finkelstein JN. Radiation induced pulmonary fibrosis as a model of progressive fibrosis: Contributions of DNA damage, inflammatory response and cellular senescence genes. Exp Lung Res 2017; 43:134-149. [PMID: 28534660 DOI: 10.1080/01902148.2017.1318975] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Purpose/Aim of Study: Studies of pulmonary fibrosis (PF) have resulted in DNA damage, inflammatory response, and cellular senescence being widely hypothesized to play a role in the progression of the disease. Utilizing these aforementioned terms, genomics databases were interrogated along with the term, "pulmonary fibrosis," to identify genes common among all 4 search terms. Findings were compared to data derived from a model of radiation-induced progressive pulmonary fibrosis (RIPF) to verify that these genes are similarly expressed, supporting the use of radiation as a model for diseases involving PF, such as human idiopathic pulmonary fibrosis (IPF). MATERIALS AND METHODS In an established model of RIPF, C57BL/6J mice were exposed to 12.5 Gy thorax irradiation and sacrificed at 24 hours, 1, 4, 12, and 32 weeks following exposure, and lung tissue was compared to age-matched controls by RNA sequencing. RESULTS Of 176 PF associated gene transcripts identified by database interrogation, 146 (>82%) were present in our experimental model, throughout the progression of RIPF. Analysis revealed that nearly 85% of PF gene transcripts were associated with at least 1 other search term. Furthermore, of 22 genes common to all four terms, 16 were present experimentally in RIPF. CONCLUSIONS This illustrates the validity of RIPF as a model of progressive PF/IPF based on the numbers of transcripts reported in both literature and observed experimentally. Well characterized genes and proteins are implicated in this model, supporting the hypotheses that DNA damage, inflammatory response and cellular senescence are associated with the pathogenesis of PF.
Collapse
Affiliation(s)
- Tyler A Beach
- a Department of Environmental Medicine , University of Rochester Medical Center , Rochester , New York , USA
| | - Carl J Johnston
- a Department of Environmental Medicine , University of Rochester Medical Center , Rochester , New York , USA.,b Department of Pediatrics and Neonatology , University of Rochester School of Medicine and Dentistry , Rochester , New York , USA
| | - Angela M Groves
- b Department of Pediatrics and Neonatology , University of Rochester School of Medicine and Dentistry , Rochester , New York , USA
| | - Jacqueline P Williams
- a Department of Environmental Medicine , University of Rochester Medical Center , Rochester , New York , USA
| | - Jacob N Finkelstein
- a Department of Environmental Medicine , University of Rochester Medical Center , Rochester , New York , USA.,b Department of Pediatrics and Neonatology , University of Rochester School of Medicine and Dentistry , Rochester , New York , USA
| |
Collapse
|