1
|
Wang Y, Zhang J, Liu Y, Jiang H, Wu B, Zhao S, Ouyang W, Hu Y, Lu B, Su S. Low-dose radiation as a potential strategy for alleviating lung injury caused by radiotherapy combined with immunotherapy: A preclinical study. Radiother Oncol 2025; 206:110823. [PMID: 39993602 DOI: 10.1016/j.radonc.2025.110823] [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: 10/16/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025]
Abstract
BACKGROUND AND PURPOSE Radiotherapy combined with immunotherapy has been shown to improve thoracic tumor outcomes while increasing the risk of lung injury. Low-dose radiotherapy (LD-RT) has been proven efficient in managing inflammatory diseases. This study aims to investigate whether LD-RT might alleviate lung injury induced by radiotherapy combined with immunotherapy, and attempt to explore its underlying mechanisms thereby offering novel insights for clinical application. METHODS To establish a mouse model of lung injury caused by radiotherapy combined with immunotherapy, C57 BL/6J mice received intraperitoneal injections of programmed death 1(PD-1) inhibitor weekly and a single dose of 15 Gy whole thoracic irradiation. Then they received a single dose of LD-RT at 1.0 Gy or 1.5 Gy on Day 14 or 28. The mice were euthanized on Day 42, and lung tissue samples were collected for HE, Masson's trichrome, and immunohistochemical staining to evaluate lung tissue damage, fibrosis, and lymphocyte infiltration. The expression levels of cytokine were quantified by the enzyme-linked immunosorbent assay. RESULTS Both low-dose of 1.0 Gy and 1.5 Gy attenuated lung injury caused by radiotherapy combined with PD-1 inhibitors, but 1.5 Gy was more effective. Compared with Day 14, LD-RT at 1.5 Gy on Day 28 was more effective in alleviating alveolar inflammation and reducing collagen deposition, and inhibiting lymphocyte infiltration and secretion of inflammatory cytokine in lung tissue. CONCLUSION Low-dose radiation alleviated lung injury caused by radiotherapy combined with PD-1 inhibitor, and the alleviating effect is closely related to the timing and dose of the radiotherapy administered.
Collapse
Affiliation(s)
- Yu Wang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Jing Zhang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Yao Liu
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Han Jiang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Bibo Wu
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Shasha Zhao
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - WeiWei Ouyang
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Yinxiang Hu
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Bing Lu
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China
| | - Shengfa Su
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, China; Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China; Teaching and Research Department of Oncology, Clinical Medical College of Guizhou Medical University, Guiyang, China.
| |
Collapse
|
2
|
Wang SY, Wu JX, An X, Yuan Z, Ren YF, Yu XF, Tian XD, Wei W. Structural and temporal dynamics analysis on immune response in low-dose radiation: History, research hotspots and emerging trends. World J Radiol 2025; 17:101636. [PMID: 40309477 PMCID: PMC12038408 DOI: 10.4329/wjr.v17.i4.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Radiotherapy (RT) is a cornerstone of cancer treatment. Compared with conventional high-dose radiation, low-dose radiation (LDR) causes less damage to normal tissues while potentially modulating immune responses and inhibiting tumor growth. LDR stimulates both innate and adaptive immunity, enhancing the activity of natural killer cells, dendritic cells, and T cells. However, the mechanisms underlying the effects of LDR on the immune system remain unclear. AIM To explore the history, research hotspots, and emerging trends in immune response to LDR literature over the past two decades. METHODS Publications on immune responses to LDR were retrieved from the Web of Science Core Collection. Bibliometric tools, including CiteSpace and HistCite, were used to identify historical features, active topics, and emerging trends in this field. RESULTS Analysis of 1244 publications over the past two decades revealed a significant surge in research on immune responses to LDR, particularly in the last decade. Key journals such as INR J Radiat Biol, Cancers, and Radiat Res published pivotal studies. Citation networks identified key studies by authors like Twyman-Saint Victor C (2015) and Vanpouille-Box C (2017). Keyword analysis revealed hotspots such as ipilimumab, stereotactic body RT, and targeted therapy, possibly identifying future research directions. Temporal variations in keyword clusters and alluvial flow maps illustrate the evolution of research themes over time. CONCLUSION This bibliometric analysis provides valuable insights into the evolution of studies on responses to LDR, highlights research trends, and identifies emerging areas for further investigation.
Collapse
Affiliation(s)
- Shu-Yuan Wang
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Jia-Xing Wu
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xian An
- Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhen Yuan
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Yi-Fan Ren
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiu-Feng Yu
- Department of General Medicine, Tuberculosis Hospital of Shaanxi Province, Xi’an 710105, Shaanxi Province, China
| | - Xiao-Dong Tian
- Department of Thoracic Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Wei Wei
- Senior Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
3
|
Zheng L, Cai W, Ke Y, Hu X, Yang C, Zhang R, Wu H, Liu D, Yu H, Wu C. Cancer‑associated fibroblasts: a pivotal regulator of tumor microenvironment in the context of radiotherapy. Cell Commun Signal 2025; 23:147. [PMID: 40114180 PMCID: PMC11927177 DOI: 10.1186/s12964-025-02138-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND In the course of tumor treatment, radiation therapy (RT) not only kills cancer cells, but also induces complex biological effects in non-malignant cells around cancer cells. These biological effects such as angiogenesis, changes in stromal composition and immune cell infiltration remodel the tumor microenvironment (TME). As one of the major components of the TME, Cancer‑associated fibroblasts (CAFs) are not only involved in tumorigenesis, progression, recurrence, and metastasis but also regulate the tumor-associated immune microenvironment. CAFs and tumor cells or immune cells have complex intercellular communication in the context of tumor radiation. MAIN CONTENT Different cellular precursors, spatial location differences, absence of specific markers, and advances in single-cell sequencing technology have gradually made the abundant heterogeneity of CAFs well known. Due to unique radioresistance properties, CAFs can survive under high doses of ionizing radiation. However, radiation can induce phenotypic and functional changes in CAFs and further act on tumor cells and immune cells to promote or inhibit tumor progression. To date, the effect of RT on CAFs and the effect of irradiated CAFs on tumor progression and TME are still not well defined. CONCLUSION In this review, we review the origin, phenotypic, and functional heterogeneity of CAFs and describe the effects of RT on CAFs, focusing on the mutual crosstalk between CAFs and tumor or immune cells after radiation. We also discuss emerging strategies for targeted CAFs therapy.
Collapse
Affiliation(s)
- Linhui Zheng
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Wenqi Cai
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Yuan Ke
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Xiaoyan Hu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chunqian Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Runze Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Huachao Wu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Dong Liu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Haijun Yu
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
- Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China.
| | - Chaoyan Wu
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Zhongnan Hospital of Wuhan University, 169, Donghu Road, Wuchang District, Wuhan, Hubei, 430071, China.
| |
Collapse
|
4
|
Yin J, Ye Y, Gao Y, Xu Q, Su M, Sun S, Xu W, Fu Q, Wang A, Hu S. Low-Dose Ionizing Radiation and Male Reproductive Immunity: Elucidating Subtle Modulations and Long-Term Health Implications. Int J Mol Sci 2025; 26:2269. [PMID: 40076897 PMCID: PMC11900348 DOI: 10.3390/ijms26052269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Low-dose ionizing radiation (LDIR) is a prevalent environmental factor with profound impacts on male reproductive health, particularly on the testicular immune microenvironment. This review examines the multifaceted effects of LDIR, emphasizing its ability to induce genotoxic stress, oxidative damage, and epigenetic modifications in reproductive cells. These alterations compromise DNA repair, disrupt chromatin structure, and induce immune dysregulation. Immune cells such as macrophages, T cells, natural killer cells, and dendritic cells exhibit significant functional changes under LDIR exposure, destabilizing the immune privilege critical for normal spermatogenesis. The long-term health implications of LDIR include impaired sperm quality, reduced fertility, and transgenerational risks through heritable genomic instability. This review underscores the importance of exploring the mechanisms underlying immune dysregulation and developing effective protective strategies. While LDIR's full impact on male reproductive health remains to be elucidated, addressing the gaps in our understanding of immune microenvironmental changes is crucial for mitigating its adverse effects and improving reproductive health outcomes.
Collapse
Affiliation(s)
- Jiacheng Yin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Yifan Ye
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Yuankai Gao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Qing Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Muzhe Su
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Shengkui Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Wenhui Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
| | - Qian Fu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - An Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| | - Sumin Hu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; (J.Y.); (Y.Y.); (Y.G.); (Q.X.); (M.S.); (S.S.); (Q.F.)
| |
Collapse
|
5
|
Lin Y, Lang H, Gao P, Miao X, Guo Q, Hao Y, Ai T, Li J, Zhang J, Guo G. Electromagnetic pulse exposure induces neuroinflammation and blood-brain barrier disruption by activating the NLRP3 inflammasome/NF-κB signaling pathway in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117972. [PMID: 40020384 DOI: 10.1016/j.ecoenv.2025.117972] [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: 11/27/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
The electromagnetic pulse (EMP) is a widespread electromagnetic disturbance that can disrupt electronic systems and pose health risks to personnel in operational areas. The biological effects of EMP radiation, especially on the central nervous system (CNS), are not yet fully understood but are gaining attention. This study examines the impact of EMP on the CNS using microglial cells as a model system. We found that mice exposed to a field strength of 600 kV/m with 1000 pulses per day for two weeks exhibited increased levels of oxidative stress. This exposure induced a microglia polarization to the M1 state, leading to neuroinflammation and disruption of the blood-brain barrier (BBB) by the pro-inflammatory response of microglia. Further analysis revealed that the NLRP3 inflammasome/NF-κB signaling pathway modulates the pro-inflammatory mechanisms of EMP irradiation. In conclusion, our findings show that EMP irradiation triggers neuroinflammation and BBB damage via NLRP3 inflammasome/NF-κB activation. This research highlights the effects of EMP radiation on the CNS and offers valuable insights into the potential targets for biomedical protection against it.
Collapse
Affiliation(s)
- Yanyun Lin
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Haiyang Lang
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China; The Ministry-of-Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Airforce Medical University, Xi'an 710032, China
| | - Peng Gao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Xia Miao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Qiyan Guo
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Yue Hao
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Tao Ai
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Jing Li
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China
| | - Jie Zhang
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China.
| | - Guozhen Guo
- Department of Radiation Medicine and Protection, Faculty of Preventive Medicine, Airforce Medical University, Xi'an 710032, China.
| |
Collapse
|
6
|
Hoveidaei A, Karimi M, Salmannezhad A, Tavakoli Y, Taghavi SP, Hoveidaei AH. Low-dose Radiation Therapy (LDRT) in Managing Osteoarthritis: A Comprehensive Review. CURRENT THERAPEUTIC RESEARCH 2025; 102:100777. [PMID: 40177366 PMCID: PMC11964493 DOI: 10.1016/j.curtheres.2025.100777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/30/2025] [Indexed: 04/05/2025]
Abstract
Osteoarthritis (OA) is the most common degenerative arthropathy, impacting the quality of life for millions worldwide. It typically presents with chronic pain, stiffness, and reduced mobility in the affected joints. Nonsurgical treatments like physiotherapy or pharmacotherapy may provide limited relief and may have adverse effects and complications. Recently, low-dose radiation therapy (LDRT) has emerged as a potential alternative for managing OA, utilizing its anti-inflammatory effects. LDRT's anti-inflammatory effects involve modulating immune responses, reducing pro-inflammatory cytokines, and inducing apoptosis in inflammatory cells. Clinical studies show varying degrees of symptom relief, with some patients experiencing pain reduction and improved joint mobility while others show minimal response. The variability in LDRT treatment designs, radiation dosages, and patient populations complicates standardized treatment protocols and raises concerns about potential carcinogenic risks. Despite these issues, LDRT shows promise as an alternative to other OA treatments, especially for patients who don't respond to other treatments. This review aims to provide updated information on the effectiveness, mechanisms, and safety of LDRT in treating OA. We reviewed the literature of studies on the safety and efficacy of LDRT on affected joints by OA, its biological effects, potential therapeutic and adverse effects, application and contraindications, clinical outcomes, and clinical evidence in subjects with OA.
Collapse
Affiliation(s)
- Armin Hoveidaei
- Students’ Scientific Research Center, Exceptional Talents Development Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kyiv, Ukraine
| | | | - Yasaman Tavakoli
- Student Research Committee, Department of Medicine, Mazandaran University of Medical Science, Sari, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, Maryland
| |
Collapse
|
7
|
Schreiner U, Huberti H. How Effective is Low-dose Radiotherapy (LD-RT) for Heberden's Osteoarthritis? An Analysis of the Current Literature. ZEITSCHRIFT FUR ORTHOPADIE UND UNFALLCHIRURGIE 2025. [PMID: 39870083 DOI: 10.1055/a-2489-5071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Low-dose radiotherapy is an established treatment option for non-malignant skeletal disorders. It is used in the treatment of Heberden's osteoarthritis (HA), but the evidence of efficacy does not seem to be certain. This paper reviews current literature for scientific evidence of efficacy in the treatment of HA.The PubMed and Cochrane Library databases were searched for relevant publications.9 publications were identified that published data from 7 studies. Only one study was randomised, placebo-controlled and double-blinded. None of the studies exclusively investigated HA. The studies were all inhomogenous with regard to inclusion criteria, follow-up criteria, radiation mode and interpretation or treatment success. In the RCT, no difference was found between the verum and control groups.The study situation is currently weak. The researched publications are not sufficiently focused on the collective of Heberden's osteoarthritis and are generally too inhomogenous with regard to the criteria applied. Future targeted studies are therefore required to prove efficacy.
Collapse
Affiliation(s)
- Ute Schreiner
- Geschäftsstelle Mannheim, Deutsche Arthrose-Hilfe e.V., Mannheim, Deutschland
| | - Helmut Huberti
- Geschäftsstelle Mannheim, Deutsche Arthrose-Hilfe e.V., Mannheim, Deutschland
| |
Collapse
|
8
|
Yamaga S, Murao A, Zhou M, Aziz M, Brenner M, Wang P. Radiation-induced eCIRP impairs macrophage bacterial phagocytosis. J Leukoc Biol 2024; 116:1072-1079. [PMID: 38920274 PMCID: PMC11531804 DOI: 10.1093/jleuko/qiae132] [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: 02/01/2024] [Revised: 04/29/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Macrophages are essential immune cells for host defense against bacterial pathogens after radiation injury. However, the role of macrophage phagocytosis in infection following radiation injury remains poorly examined. Extracellular cold-inducible RNA-binding protein is a damage-associated molecular pattern that dysregulates host immune system responses such as phagocytosis. We hypothesized that radiation-induced extracellular cold-inducible RNA-binding protein release impairs macrophage phagocytosis of bacteria. Adult healthy mice were exposed to 6.5 Gy total body irradiation. Primary peritoneal macrophages isolated from adult healthy mice were exposed to 6.5 Gy radiation. Extracellular cold-inducible RNA-binding protein-neutralizing monoclonal antibody was added to the cell culture prior to irradiation. Bacterial phagocytosis by peritoneal macrophages was assessed using pHrodo Green-labeled Escherichia coli 7 d after irradiation ex vivo and in vitro. Bacterial phagocytosis was also assessed after treatment with recombinant murine cold-inducible RNA-binding protein. Rac1 and ARP2 protein expression in cell lysates and extracellular cold-inducible RNA-binding protein levels in the peritoneal lavage were assessed by western blotting. Bacterial phagocytosis by peritoneal macrophages was significantly decreased after irradiation compared with controls ex vivo and in vitro. Rac1 and ARP2 expression in the peritoneal macrophages were downregulated after total body irradiation. Total body irradiation significantly increased extracellular cold-inducible RNA-binding protein levels in the peritoneal cavity. Recombinant murine cold-inducible RNA-binding protein significantly decreased bacterial phagocytosis in a dose-dependent manner. Extracellular cold-inducible RNA-binding protein monoclonal antibody restored bacterial phagocytosis by peritoneal macrophages after irradiation. Ionizing radiation exposure impairs bacterial phagocytosis by macrophages after irradiation. Neutralization of extracellular cold-inducible RNA-binding protein restores the phagocytic ability of macrophages after irradiation. Our findings elucidate a novel mechanism of immune dysfunction and provide a potential new therapeutic approach for limiting infection after radiation injury.
Collapse
Affiliation(s)
- Satoshi Yamaga
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Mian Zhou
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| | - Max Brenner
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Dr., Manhasset, NY 11030, United States
| |
Collapse
|
9
|
Richard SA, Roy SK, Asiamah EA. Pivotal Role of Cranial Irradiation-Induced Peripheral, Intrinsic, and Brain-Engrafting Macrophages in Malignant Glioma. Clin Med Insights Oncol 2024; 18:11795549241282098. [PMID: 39421649 PMCID: PMC11483687 DOI: 10.1177/11795549241282098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024] Open
Abstract
Malignant (high-grade) gliomas are aggressive intrinsic brain tumors that diffusely infiltrate the brain parenchyma. They comprise of World Health Organization (WHO) grade III and IV gliomas. Ionizing radiation or irradiation (IR) is frequently utilized in the treatment of both primary as well as metastatic brain tumors. On the contrary, macrophages (MΦ) are the most copious infiltrating immune cells of all the different cell types colonizing glioma. MΦ at tumor milieu are referred to as tumor-associated macrophages (TAMΦ). In malignant gliomas milieu, TAMΦ are also polarized into two distinct phenotypes such as M1 TAMΦ or M2 TAMΦ, which are capable of inhibiting or promoting tumor growth, respectively. Cranial-IR such as x- and γ-IR are sufficient to induce the migration of peripherally derived MΦ into the brain parenchyma. The IR facilitate a more immunosuppressive milieu via the stimulation of efferocytosis in TAMΦ, and an upsurge of tumor cell engulfment by TAMΦ exhibited detrimental effect of the anti-tumoral immune response in glioma. The MΦ inside the tumor mass are associated with multiple phenomena that include IR resistance and enrichment of the M2 MΦ after IR is able to facilitate glioblastoma multiforme (GBM) recurrence. Reviews on the role of cranial IR-induced peripheral and brain-engrafting macrophages (BeMΦ) in glioma are lacking. Specifically, most studies on peripheral, intrinsic as well as beMΦ on IR focus on WHO grade III and IV. Thus, this review precisely focuses primary on WHO grade III as well as IV gliomas.
Collapse
Affiliation(s)
- Seidu A Richard
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), UK, Ghana
- Institute of Neuroscience, Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Sagor Kumar Roy
- Department of Neurology, TMSS Medical College and Hospital, Bogura, Bangladesh
| | - Emmanuel Akomanin Asiamah
- Department of Medical Laboratory Sciences, School of Allied Health Sciences, University of Health and Allied Sciences, Ho, Ghana
| |
Collapse
|
10
|
Sheva K, Roy Chowdhury S, Kravchenko-Balasha N, Meirovitz A. Molecular Changes in Breast Cancer Induced by Radiation Therapy. Int J Radiat Oncol Biol Phys 2024; 120:465-481. [PMID: 38508467 DOI: 10.1016/j.ijrobp.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Breast cancer treatments are based on prognostic clinicopathologic features that form the basis for therapeutic guidelines. Although the utilization of these guidelines has decreased breast cancer-associated mortality rates over the past three decades, they are not adequate for individualized therapy. Radiation therapy (RT) is the backbone of breast cancer treatment. Although a highly successful therapeutic modality clinically, from a biological perspective, preclinical studies have shown RT to have the potential to alter tumor cell phenotype, immunogenicity, and the surrounding microenvironment, potentially changing the behavior of cancer cells and resulting in a significant variation in RT response. This review presents the recent advances in revealing the complex molecular changes induced by RT in the treatment of breast cancer and highlights the complexities of translating this information into clinically relevant tools for improved prognostic insights and the revelation of novel approaches for optimizing RT. METHODS AND MATERIALS Current literature was reviewed with a focus on recent advances made in the elucidation of tumor-associated radiation-induced molecular changes across molecular, genetic, and proteomic bases. This review was structured with the aim of providing an up-to-date overview over the very broad and complex subject matter of radiation-induced molecular changes and radioresistance, familiarizing the reader with the broader issue at hand. RESULTS The subject of radiation-induced molecular changes in breast cancer has been broached from various physiological focal points including that of the immune system, immunogenicity and the abscopal effect, tumor hypoxia, breast cancer classification and subtyping, molecular heterogeneity, and molecular plasticity. It is becoming increasingly apparent that breast cancer clinical subtyping alone does not adequately account for variation in RT response or radioresistance. Multiple components of the tumor microenvironment and immune system, delivered RT dose and fractionation schedules, radiation-induced bystander effects, and intrinsic tumor physiology and heterogeneity all contribute to the resultant RT outcome. CONCLUSIONS Despite recent advances and improvements in anticancer therapies, tumor resistance remains a significant challenge. As new analytical techniques and technologies continue to provide crucial insight into the complex molecular mechanisms of breast cancer and its treatment responses, it is becoming more evident that personalized anticancer treatment regimens may be vital in overcoming radioresistance.
Collapse
Affiliation(s)
- Kim Sheva
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| | - Sangita Roy Chowdhury
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Amichay Meirovitz
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| |
Collapse
|
11
|
Wang SW, Zheng QY, Hong WF, Tang BF, Hsu SJ, Zhang Y, Zheng XB, Zeng ZC, Gao C, Ke AW, Du SS. Mechanism of immune activation mediated by genomic instability and its implication in radiotherapy combined with immune checkpoint inhibitors. Radiother Oncol 2024; 199:110424. [PMID: 38997092 DOI: 10.1016/j.radonc.2024.110424] [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: 04/07/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Various genetic and epigenetic changes associated with genomic instability (GI), including DNA damage repair defects, chromosomal instability, and mitochondrial GI, contribute to development and progression of cancer. These alterations not only result in DNA leakage into the cytoplasm, either directly or through micronuclei, but also trigger downstream inflammatory signals, such as the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway. Apart from directly inducing DNA damage to eliminate cancer cells, radiotherapy (RT) exerts its antitumor effects through intracellular DNA damage sensing mechanisms, leading to the activation of downstream inflammatory signaling pathways. This not only enables local tumor control but also reshapes the immune microenvironment, triggering systemic immune responses. The combination of RT and immunotherapy has emerged as a promising approach to increase the probability of abscopal effects, where distant tumors respond to treatment due to the systemic immunomodulatory effects. This review emphasizes the importance of GI in cancer biology and elucidates the mechanisms by which RT induces GI remodeling of the immune microenvironment. By elucidating the mechanisms of GI and RT-induced immune responses, we aim to emphasize the crucial importance of this approach in modern oncology. Understanding the impact of GI on tumor biological behavior and therapeutic response, as well as the possibility of activating systemic anti-tumor immunity through RT, will pave the way for the development of new treatment strategies and improve prognosis for patients.
Collapse
Affiliation(s)
- Si-Wei Wang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China
| | - Qiu-Yi Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Wei-Feng Hong
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Bu-Fu Tang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Shu-Jung Hsu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Yang Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Xiao-Bin Zheng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Zhao-Chong Zeng
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China
| | - Chao Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Ai-Wu Ke
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Liver Cancer Institute, Fudan University, Shanghai 200030, China.
| | - Shi-Suo Du
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai 200030, China.
| |
Collapse
|
12
|
Shao Y, Han S, Hou Z, Yang C, Zhao Y. Tumor-associated macrophages within the immunological milieu: An emerging focal point for therapeutic intervention. Heliyon 2024; 10:e36839. [PMID: 39281573 PMCID: PMC11401039 DOI: 10.1016/j.heliyon.2024.e36839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Tumor-associated macrophages play an important role in the tumor immune microenvironment, and regulating the function of tumor-associated macrophages has important therapeutic potential in tumor therapy. Mature macrophages could migrate to the tumor microenvironment, influencing multiple factors such as tumor cell proliferation, invasion, metastasis, extracellular matrix remodeling, immune suppression, and drug resistance. As a major component of the tumor microenvironment, tumor-associated macrophages crosstalk with other immune cells. Currently, tumor-associated macrophages have garnered considerable attention in tumor therapy, broadening the spectrum of drug selection to some extent, thereby aiding in mitigating the prevailing clinical drug resistance dilemma. This article summarizes the recent advances in tumor-associated macrophages concerning immunology, drug targeting mechanisms for tumor-associated macrophages treatment, new developments, and existing challenges, offering insights for future therapeutic approaches. In addition, this paper summarized the impact of tumor-associated macrophages on current clinical therapies, discussed the advantages and disadvantages of targeted tumor-associated macrophages therapy compared with existing tumor therapies, and predicted and discussed the future role of targeted tumor-associated macrophages therapy and the issues that need to be focused on.
Collapse
Affiliation(s)
- Yanchi Shao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Song Han
- The First Hospital of Jilin University, Changchun, China
| | - Zhenxin Hou
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Chen Yang
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yanbin Zhao
- Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
13
|
Srinivasan D, Subbarayan R, Srivastava N, Radhakrishnan A, Adtani PN, Chauhan A, Krishnamoorthy L. A comprehensive overview of radiation therapy impacts of various cancer treatments and pivotal role in the immune system. Cell Biochem Funct 2024; 42:e4103. [PMID: 39073207 DOI: 10.1002/cbf.4103] [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: 05/13/2024] [Revised: 06/25/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024]
Abstract
The cancer treatment landscape is significantly evolving, focusing on advanced radiation therapy methods to maximize effectiveness and minimize the adverse effects. Recognized as a pivotal component in cancer and disease treatment, radiation therapy (RT) has drawn attention in recent research that delves into its intricate interplay with inflammation and the immune response. This exploration unveils the underlying processes that significantly influence treatment outcomes. In this context, the potential advantages of combining bronchoscopy with RT across diverse clinical scenarios, alongside the targeted impact of brachytherapy, are explored. Concurrently, radiation treatments serve multifaceted roles such as DNA repair, cell elimination, and generating immune stress signaling molecules known as damage-associated molecular patterns, elucidating their effectiveness in treating various diseases. External beam RT introduces versatility by utilizing particles such as photons, electrons, protons, or carbon ions, each offering distinct advantages. Advanced RT techniques contribute to the evolving landscape, with emerging technologies like FLASH, spatially fractionated RT, and others poised to revolutionize the field. The comprehension of RT, striving for improved treatment outcomes, reduced side effects, and facilitating personalized and innovative treatments for cancer and noncancer patients. After navigating these advancements, the goal is fixed to usher in a new era in which RT is a cornerstone of precision and effectiveness in medical interventions. In summarizing the myriad findings, the review underscores the significance of understanding the differential impacts of radiation approaches on inflammation and immune modulation, offering valuable insights for developing innovative therapeutic interventions that harness the immune system in conjunction with RT.
Collapse
Affiliation(s)
- Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, Faculty of Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Arunkumar Radhakrishnan
- Department of Pharmacology, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Pooja Narain Adtani
- Department of Basic Medical and Dental Sciences, College of Dentistry, Gulf Medical University, Ajman, United Arab Emirates
| | - Ankush Chauhan
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Loganathan Krishnamoorthy
- Department of Allied Health Sciences-FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| |
Collapse
|
14
|
Rey N, Ebrahimian T, Gloaguen C, Kereselidze D, Christelle E, Brizais C, Bachelot F, Riazi G, Monceau V, Demarquay C, Zineddine IG, Klokov D, Lehoux S, Ebrahimian TG. Low to moderate dose 137Cs (γ) radiation promotes M2 type macrophage skewing and reduces atherosclerotic plaque CD68+ cell content in ApoE (-/-) mice. Sci Rep 2024; 14:12450. [PMID: 38816571 PMCID: PMC11139881 DOI: 10.1038/s41598-024-63084-x] [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: 02/12/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024] Open
Abstract
The effects of low doses of ionizing radiation on atherosclerosis remain uncertain, particularly as regards the generation of pro- or anti-inflammatory responses, and the time scale at which such effects can occur following irradiation. To explore these phenomena, we exposed atheroprone ApoE(-/-) mice to a single dose of 0, 0.05, 0.5 or 1 Gy of 137Cs (γ) administered at a 10.35 mGy min-1 dose rate and evaluated short-term (1-10 days) and long-term consequences (100 days). Bone marrow-derived macrophages were derived from mice 1 day after exposure. Irradiation was associated with a significant skewing of M0 and M2 polarized macrophages towards the M2 phenotype, as demonstrated by an increased mRNA expression of Retnla, Arg1, and Chil3 in cells from mice exposed to 0.5 or 1 Gy compared with non-irradiated animals. Minimal effects were noted in M1 cells or M1 marker mRNA. Concurrently, we observed a reduced secretion of IL-1β but enhanced IL-10 release from M0 and M2 macrophages. Effects of irradiation on circulating monocytes were most marked at day 10 post-exposure, when the 1 Gy dose was associated with enhanced numbers of both Ly6CHigh and Ly6Low cells. By day 100, levels of circulating monocytes in irradiated and non-irradiated mice were equivalent, but anti-inflammatory Ly6CLow monocytes were significantly increased in the spleen of mice exposed to 0.05 or 1 Gy. Long term exposures did not affect atherosclerotic plaque size or lipid content, as determined by Oil red O staining, whatever the dose applied. Similarly, irradiation did not affect atherosclerotic plaque collagen or smooth muscle cell content. However, we found that lesion CD68+ cell content tended to decrease with rising doses of radioactivity exposure, culminating in a significant reduction of plaque macrophage content at 1 Gy. Taken together, our results show that short- and long-term exposures to low to moderate doses of ionizing radiation drive an anti-inflammatory response, skewing bone marrow-derived macrophages towards an IL-10-secreting M2 phenotype and decreasing plaque macrophage content. These results suggest a low-grade athero-protective effect of low and moderate doses of ionizing radiation.
Collapse
Affiliation(s)
- N Rey
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - T Ebrahimian
- Department of Medicine, Lady Davis Institute for Biomedical Research, McGill University, Montreal, Canada
| | - C Gloaguen
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - D Kereselidze
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - E Christelle
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - C Brizais
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - F Bachelot
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - G Riazi
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - V Monceau
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - C Demarquay
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - I Garali Zineddine
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - D Klokov
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France
| | - S Lehoux
- Department of Medicine, Lady Davis Institute for Biomedical Research, McGill University, Montreal, Canada.
| | - Teni G Ebrahimian
- Institut de Radioprotection et de Sûreté Nucléaire, Laboratoire de Radiotoxicologie et de Radiobiologie Expérimentale, 92262, Fontenay-Aux Roses, France.
| |
Collapse
|
15
|
Bergerud KMB, Berkseth M, Pardoll DM, Ganguly S, Kleinberg LR, Lawrence J, Odde DJ, Largaespada DA, Terezakis SA, Sloan L. Radiation Therapy and Myeloid-Derived Suppressor Cells: Breaking Down Their Cancerous Partnership. Int J Radiat Oncol Biol Phys 2024; 119:42-55. [PMID: 38042450 PMCID: PMC11082936 DOI: 10.1016/j.ijrobp.2023.11.050] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
Radiation therapy (RT) has been a primary treatment modality in cancer for decades. Increasing evidence suggests that RT can induce an immunosuppressive shift via upregulation of cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). MDSCs inhibit antitumor immunity through potent immunosuppressive mechanisms and have the potential to be crucial tools for cancer prognosis and treatment. MDSCs interact with many different pathways, desensitizing tumor tissue and interacting with tumor cells to promote therapeutic resistance. Vascular damage induced by RT triggers an inflammatory signaling cascade and potentiates hypoxia in the tumor microenvironment (TME). RT can also drastically modify cytokine and chemokine signaling in the TME to promote the accumulation of MDSCs. RT activation of the cGAS-STING cytosolic DNA sensing pathway recruits MDSCs through a CCR2-mediated mechanism, inhibiting the production of type 1 interferons and hampering antitumor activity and immune surveillance in the TME. The upregulation of hypoxia-inducible factor-1 and vascular endothelial growth factor mobilizes MDSCs to the TME. After recruitment, MDSCs promote immunosuppression by releasing reactive oxygen species and upregulating nitric oxide production through inducible nitric oxide synthase expression to inhibit cytotoxic activity. Overexpression of arginase-1 on subsets of MDSCs degrades L-arginine and downregulates CD3ζ, inhibiting T-cell receptor reactivity. This review explains how radiation promotes tumor resistance through activation of immunosuppressive MDSCs in the TME and discusses current research targeting MDSCs, which could serve as a promising clinical treatment strategy in the future.
Collapse
Affiliation(s)
| | - Matthew Berkseth
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sudipto Ganguly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lawrence R Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota, St. Paul, Minnesota
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota
| | | | - Lindsey Sloan
- Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
16
|
Innuan P, Sirikul C, Anukul N, Rolin G, Dechsupa N, Kantapan J. Identifying transcriptomic profiles of iron-quercetin complex treated peripheral blood mononuclear cells from healthy volunteers and diabetic patients. Sci Rep 2024; 14:9441. [PMID: 38658734 PMCID: PMC11043337 DOI: 10.1038/s41598-024-60197-1] [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: 11/03/2023] [Accepted: 04/19/2024] [Indexed: 04/26/2024] Open
Abstract
Peripheral blood is an alternative source of stem/progenitor cells for regenerative medicine owing to its ease of retrieval and blood bank storage. Previous in vitro studies indicated that the conditioned medium derived from peripheral blood mononuclear cells (PBMCs) treated with the iron-quercetin complex (IronQ) contains potent angiogenesis and wound-healing properties. This study aims to unveil the intricate regulatory mechanisms governing the effects of IronQ on the transcriptome profiles of human PBMCs from healthy volunteers and those with diabetes mellitus (DM) using RNA sequencing analysis. Our findings revealed 3741 and 2204 differentially expressed genes (DEGs) when treating healthy and DM PBMCs with IronQ, respectively. Functional enrichment analyses underscored the biological processes shared by the DEGs in both conditions, including inflammatory responses, cell migration, cellular stress responses, and angiogenesis. A comprehensive exploration of these molecular alterations exposed a network of 20 hub genes essential in response to stimuli, cell migration, immune processes, and the mitogen-activated protein kinase (MAPK) pathway. The activation of these pathways enabled PBMCs to potentiate angiogenesis and tissue repair. Corroborating this, quantitative real-time polymerase chain reaction (qRT-PCR) and cell phenotyping confirmed the upregulation of candidate genes associated with anti-inflammatory, pro-angiogenesis, and tissue repair processes in IronQ-treated PBMCs. In summary, combining IronQ and PBMCs brings about substantial shifts in gene expression profiles and activates pathways that are crucial for tissue repair and immune response, which is promising for the enhancement of the therapeutic potential of PBMCs, especially in diabetic wound healing.
Collapse
Affiliation(s)
- Phattarawadee Innuan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chonticha Sirikul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nampeung Anukul
- Division of Transfusion Science, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Gwenaël Rolin
- INSERM CIC-1431, CHU Besançon, 25000, Besançon, France
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
17
|
Idel C, Fleckner J, Plötze-Martin K, Werner L, Rades D, Theodoraki MN, Hofmann L, Huber D, Leichtle A, Hoffmann TK, Bruchhage KL, Pries R. Partial recovery of peripheral blood monocyte subsets in head and neck squamous cell carcinoma patients upon radio(chemo)therapy is associated with decreased plasma CXCL11. BMC Cancer 2024; 24:459. [PMID: 38609887 PMCID: PMC11015641 DOI: 10.1186/s12885-024-12177-x] [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: 01/24/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) represents a common and heterogeneous malignancy of the oral cavity, pharynx and larynx. Surgery and radio(chemo)therapy are the standard treatment options and also have great influence on the composition of the tumor microenvironment and immune cell functions. However, the impact of radio(chemo)therapy on the distribution and characteristics of circulating monocyte subsets in HNSCC are not fully understood. METHODS Expression patterns of adhesion molecules and chemokine receptors CD11a (integrin-α L; LFA-1), CD11b (integrin-α M; Mac-1), CD11c (integrin-α X), CX3CR1 (CX3CL1 receptor) and checkpoint molecule PD-L1 (programmed cell death ligand-1) were investigated upon radio(chemo)therapeutic treatment using flow cytometry. Furthermore, comprehensive analysis of plasma cytokines was performed before and after treatment using ELISA measurements. RESULTS Our data reveal a partial recovery of circulating monocytes in HNSCC patients upon radio(chemo)therapeutic treatment, with differential effects of the individual therapy regimen. PD-L1 expression on non-classical monocytes significantly correlates with the individual plasma levels of chemokine CXCL11 (C-X-C motif chemokine 11). CONCLUSIONS Further comprehensive investigations on larger patient cohorts are required to elucidate the meaningfulness of peripheral blood monocyte subsets and chemokine CXCL11 as potential bioliquid indicators in HNSCC with regard to therapy response and the individual immunological situation.
Collapse
Affiliation(s)
- Christian Idel
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Jonas Fleckner
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Kirstin Plötze-Martin
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Lotte Werner
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Dirk Rades
- Department of Radiation Oncology, University of Luebeck, Luebeck, 23538, Germany
| | - Marie-Nicole Theodoraki
- Department of Otorhinolaryngology and Head & Neck Surgery, Ulm University Medical Center, Ulm, 89075, Germany
- Department of Otorhinolaryngology, Technical University Munich, Munich, Germany
| | - Linda Hofmann
- Department of Otorhinolaryngology and Head & Neck Surgery, Ulm University Medical Center, Ulm, 89075, Germany
| | - Diana Huber
- Department of Otorhinolaryngology and Head & Neck Surgery, Ulm University Medical Center, Ulm, 89075, Germany
| | - Anke Leichtle
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Thomas K Hoffmann
- Department of Otorhinolaryngology and Head & Neck Surgery, Ulm University Medical Center, Ulm, 89075, Germany
| | - Karl-Ludwig Bruchhage
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany
| | - Ralph Pries
- Department of Otorhinolaryngology and Head & Neck Surgery, University of Luebeck, Luebeck, 23538, Germany.
| |
Collapse
|
18
|
Donaubauer AJ, Becker I, Klein G, Voll RE, Weikl L, Klieser M, Barzangi S, Zhou JG, Fietkau R, Gaipl US, Frey B. Effects of serial radon spa therapy on pain and peripheral immune status in patients suffering from musculoskeletal disorders- results from a prospective, randomized, placebo-controlled trial. Front Immunol 2024; 15:1307769. [PMID: 38380316 PMCID: PMC10876773 DOI: 10.3389/fimmu.2024.1307769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/19/2024] [Indexed: 02/22/2024] Open
Abstract
In this randomized, placebo-controlled cross-over trial we aimed to investigate if radon spa therapy exerts more pain relief than exposure to warm water alone. In addition, immunological parameters were assessed in both treatment groups. In the RAD-ON02 trial, 116 patients suffering from musculoskeletal disorders (MSDs) received either serial radon spa or solely warm water baths. Pain intensity was assessed by determination of different pain parameters on a visual analogue scale and by pressure point dolorimetry at baseline and at weeks 4, 12 and 24. The longitudinal immune status of the patients was analyzed by a flow cytometry-based assay from peripheral blood at the time points of pain assessments. There were no side effects attributable to radon exposure observed. However, radon spa was superior to warm water applications at week 4 in terms of pain reduction. Pain and morning stiffness at the time of assessment were significantly reduced after radon spa (p<0.001, p<0.01) but not after warm water baths. The dolorimetry resulted in a significantly higher exerted pressure strength in patients after radon spa (p<0.001), but not after warm water applications. During the long-term follow-up, both treatment modalities reduced pain to a similar degree and pain modulation was not distorted by the participants' intake of analgesics. No significant changes in the immune status attributable specifically to radon were found, even though the increase in regulatory T cell counts occurs earlier after radon baths than after sole warm water baths and a higher level of significance is reached after radon spa at week 24. Serial radon spa has additive pain-relieving effects. The immunological parameters assessed in our study appear not to be directly linked to the pain reduction caused by radon exposure, at least in MSD patients with predominantly degenerative diseases. Clinical trial registration https://www.clinicaltrialsregister.eu/ctr-search/search?query=rad-on02, identifier 2016-002085-31; https://drks.de/search/de/trial, identifier DRKS00016019.
Collapse
Affiliation(s)
- Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ina Becker
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Gerhart Klein
- Association for Radon Spa Research and Medical Practice for Cardiology, Bad Steben, Germany
| | - Reinhard E. Voll
- Department of Rheumatology and Clinical Immunology, Medical Centre – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lena Weikl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Martin Klieser
- Association for Radon Spa Research and Medical Practice for Cardiology, Bad Steben, Germany
| | - Shakar Barzangi
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jian-Guo Zhou
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
19
|
Au NPB, Wu T, Kumar G, Jin Y, Li YYT, Chan SL, Lai JHC, Chan KWY, Yu KN, Wang X, Ma CHE. Low-dose ionizing radiation promotes motor recovery and brain rewiring by resolving inflammatory response after brain injury and stroke. Brain Behav Immun 2024; 115:43-63. [PMID: 37774892 DOI: 10.1016/j.bbi.2023.09.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/24/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023] Open
Abstract
Traumatic brain injury (TBI) and stroke share a common pathophysiology that worsens over time due to secondary tissue injury caused by sustained inflammatory response. However, studies on pharmacological interventions targeting the complex secondary injury cascade have failed to show efficacy. Here, we demonstrated that low-dose ionizing radiation (LDIR) reduced lesion size and reversed motor deficits after TBI and photothrombotic stroke. Magnetic resonance imaging demonstrated significant reduction of infarct volume in LDIR-treated mice after stroke. Systems-level transcriptomic analysis showed that genes upregulated in LDIR-treated stoke mice were enriched in pathways associated with inflammatory and immune response involving microglia. LDIR induced upregulation of anti-inflammatory- and phagocytosis-related genes, and downregulation of key pro-inflammatory cytokine production. These findings were validated by live-cell assays, in which microglia exhibited higher chemotactic and phagocytic capacities after LDIR. We observed substantial microglial clustering at the injury site, glial scar clearance and reversal of motor deficits after stroke. Cortical microglia/macrophages depletion completely abolished the beneficial effect of LDIR on motor function recovery in stroke mice. LDIR promoted axonal projections (brain rewiring) in motor cortex and recovery of brain activity detected by electroencephalography recordings months after stroke. LDIR treatment delayed by 8 h post-injury still maintained full therapeutic effects on motor recovery, indicating that LDIR is a promising therapeutic strategy for TBI and stroke.
Collapse
Affiliation(s)
| | - Tan Wu
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Yuting Jin
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | | | - Shun Lam Chan
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China
| | - Joseph Ho Chi Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Kannie Wai Yan Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| | - Kwan Ngok Yu
- Department of Physics, City University of Hong Kong, Hong Kong, China
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; Department of Surgery, Chinese University of Hong Kong, Hong Kong, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Hong Kong, China; City University of Hong Kong Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
20
|
Singh D, Malhotra P, Agarwal P, Kumar R. N-acetyl-l-tryptophan (NAT) ameliorates radiation-induced cell death in murine macrophages J774A.1 via regulating redox homeostasis and mitochondrial dysfunction. J Biochem Mol Toxicol 2024; 38:e23529. [PMID: 37702290 DOI: 10.1002/jbt.23529] [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: 02/21/2023] [Revised: 08/02/2023] [Accepted: 08/25/2023] [Indexed: 09/14/2023]
Abstract
Ionizing radiation interacts with the immune system and induces molecular damage in the cellular milieu by generating reactive oxygen species (ROS) leading to cell death. The present study was performed to investigate the protective efficacy of N-acetyl-L-tryptophan (NAT) against gamma-radiation-induced cell death in murine macrophage J774A.1 cells. The radioprotective efficacy of NAT was evaluated in terms of cell survivability, effect on antioxidant enzyme activity, and free radicals inhibition. Radioprotective efficacy of NAT pretreatment to irradiated cells was assessed via cell cycle progression, mitochondrial membrane potential (MMP) perturbation, and apoptosis regulation using flow cytometry. Results of the study demonstrated significant radioprotective efficacy (>80%) of NAT in irradiated cells as estimated by sulforhodamine B (SRB), MTT, and clonogenic assay. Significant (p < 0.001) reduction in ROS, xanthine oxidase, and mitochondrial superoxide levels along with increment in catalase, glutathione-s-transferase, glutathione, and ATPase activities in NAT pretreated plus irradiated cells was observed as compared to the gamma-irradiated cells. Further, significant (p < 0.001) stabilization of MMP and reduction in apoptosis was also observed in NAT pretreated plus irradiated cells as compared to irradiated cells that not pretreated with NAT. The current study demonstrates that NAT pretreatment to irradiated cells protects against gamma radiation-induced cell death by reducing oxidative stress, stabilizing MMP, and inhibiting apoptosis. These observations conclusively highlight the potential of developing NAT as a prospective radioprotective agent upon further validation using in-depth preclinical assessment in cellular and animal models.
Collapse
Affiliation(s)
- Darshana Singh
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Poonam Malhotra
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Prerna Agarwal
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Raj Kumar
- Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| |
Collapse
|
21
|
Dinakar K, Jakka MK, Vemannagari PKR, Mohan A, Subramanian BV, Bodagala VD, Bhuma V, Das P, Bonala SR, Vutukuru VR. Efficacy of low-dose lung radiotherapy in the management of COVID-19 patients: a randomised, open-label study. Br J Radiol 2023; 96:20230022. [PMID: 37751170 PMCID: PMC10646638 DOI: 10.1259/bjr.20230022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVE Evaluate role of low-dose radiotherapy (LDRT) in COVID-19 pneumonia. METHODS Sixty-five patients 40 years or older tested positive for COVID-19 reverse transcriptase-polymerase chain reaction with mild to moderate acute respiratory distress syndrome (ARDS), were randomised 1:1, from 4 June 2021, to either best standard of care (control arm) according to the Indian Council of Medical Research guidelines or a single dose of LDRT (LDRT-0.5Gy) to both lungs along with best standard of care (experimental arm). The primary outcome was either progression to severe disease (PaO2/FiO2 ratio <100 mmHg) within 28 days of randomisation or all-cause mortality at 28 days. If the primary outcome could have been prevented, it was considered "favourable"; if not, it was considered "unfavourable." RESULTS Thirty-three patients were allocated to experimental arm, 32 to control arm. An intention to treat analysis was performed. Unfavourable outcome was seen in 5 (15.2%) patients in experimental arm, vs , 12 (37.5%) patients in control arm, odds of an unfavourable outcome in experimental arm were 0.3, 95% CI 0.09-0.97; two-sided p = 0.04. Four and five patients died in experimental and control arm, respectively. No radiation-induced toxicity was observed. CONCLUSION LDRT reduced the number of patients with unfavourable outcome at 28 days. ADVANCES IN KNOWLEDGE One of the few randomised studies showing reduced unfavourable outcome in mild to moderate ARDS COVID-19 patients receiving LDRT.CTRI/2021/06/034001, Clinical Trials Registry - India (ICMR-NIMS).
Collapse
Affiliation(s)
- Kootala Dinakar
- Department of Radiation Oncology, Government Medical College, Ananthapur, Andhra Pradesh, India
| | - Mohan Krishna Jakka
- Department of Radiation Oncology, Sri Venkateswara Institute of Medical Sciences, SVIMS Cancer Centre, Tirupati, Andhra Pradesh, India
| | - Pavan Kumar Reddy Vemannagari
- Department of Radiation Oncology, Sri Venkateswara Institute of Medical Sciences, SVIMS Cancer Centre, Tirupati, Andhra Pradesh, India
| | - Alladi Mohan
- Department of Medicine, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Bala Venkat Subramanian
- Department of Radiation Oncology, Sri Venkateswara Institute of Medical Sciences, SVIMS Cancer Centre, Tirupati, Andhra Pradesh, India
| | | | - Vengamma Bhuma
- Department of Neurology, Sri Venkateswara Institute of Medical Sciences, Tirupati, Andhra Pradesh, India
| | - Pranabandhu Das
- Department of Radiation Oncology, Sri Venkateswara Institute of Medical Sciences, SVIMS Cancer Centre, Tirupati, Andhra Pradesh, India
| | - Sreenivasa Rao Bonala
- Department of Radiation Oncology, Sri Venkateswara Institute of Medical Sciences, SVIMS Cancer Centre, Tirupati, Andhra Pradesh, India
| | | |
Collapse
|
22
|
Weissmann T, Rückert M, Putz F, Donaubauer AJ, Hecht M, Schnellhardt S, Schubert P, Roesch J, Höfler D, Ott OJ, Haderlein M, Lettmaier S, Fietkau R, Frey B, Gaipl US, Deloch L. Low-dose radiotherapy of osteoarthritis: from biological findings to clinical effects-challenges for future studies. Strahlenther Onkol 2023; 199:1164-1172. [PMID: 36602569 PMCID: PMC10674008 DOI: 10.1007/s00066-022-02038-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/25/2022] [Indexed: 01/06/2023]
Abstract
Osteoarthritis (OA) is one of the most common and socioeconomically relevant diseases, with rising incidence and prevalence especially with regard to an ageing population in the Western world. Over the decades, the scientific perception of OA has shifted from a simple degeneration of cartilage and bone to a multifactorial disease involving various cell types and immunomodulatory factors. Despite a wide range of conventional treatment modalities available, a significant proportion of patients remain treatment refractory. Low-dose radiotherapy (LDRT) has been used for decades in the treatment of patients with inflammatory and/or degenerative diseases and has proven a viable option even in cohorts of patients with a rather poor prognosis. While its justification mainly derives from a vast body of empirical evidence, prospective randomized trials have until now failed to prove the effectiveness of LDRT. Nevertheless, over the decades, adaptions of LDRT treatment modalities have evolved using lower dosages with establishment of different treatment schedules for which definitive clinical proof is still pending. Preclinical research has revealed that the immune system is modulated by LDRT and very recently osteoimmunological mechanisms have been described. Future studies and investigations further elucidating the underlying mechanisms are an essential key to clarify the optimal patient stratification and treatment procedure, considering the patients' inflammatory status, age, and sex. The present review aims not only to present clinical and preclinical knowledge about the mechanistic and beneficial effects of LDRT, but also to emphasize topics that will need to be addressed in future studies. Further, a concise overview of the current status of the underlying radiobiological knowledge of LDRT for clinicians is given, while seeking to stimulate further translational research.
Collapse
Affiliation(s)
- Thomas Weissmann
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Florian Putz
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Anna-Jasmina Donaubauer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Markus Hecht
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Sören Schnellhardt
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Philipp Schubert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Johannes Roesch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Daniel Höfler
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Oliver J Ott
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Marlen Haderlein
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Sebastian Lettmaier
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
23
|
Pereira F, Petroianu A, Geraldo JM, Pereira C. Megavoltage radiotherapy effects on organs of the reticuloendothelial system. Acta Cir Bras 2023; 38:e384123. [PMID: 37878983 PMCID: PMC10592569 DOI: 10.1590/acb384123] [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: 05/28/2023] [Accepted: 07/30/2023] [Indexed: 10/27/2023] Open
Abstract
PURPOSE To study the uptake capacity of cells from the reticuloendothelial system after irradiation with high-energy X-rays. METHODS Eighteen male Wistar rats were distributed in three groups: group A (n = 6): control, unirradiated animals studied alongside animals from group B; group B (n = 6) and group C (n = 6): animals irradiated and studied after 24 and 48 hours, respectively. The rats were anesthetized and placed on a 10 MV linear accelerator. Next, they were irradiated in the abdominal region, with 8 Gy. Twenty-four (groups A and B) and 48 hours later (group C), a colloidal carbon solution (1 mL/kg) was intravenously injected in the tail vein. Fifty minutes later, the spleens and livers were withdrawn and prepared to be studied. Kupffer cells and splenic macrophages containing carbon pigments were counted in an optical microscope. Arithmetic means were calculated for each group and compared among them. RESULTS X-rays were associated with a reduced number of Kupffer cells containing colloidal carbon, proliferation and enlargement of biliary ducts, hypoplasia, and hepatocyte necrosis. In the irradiated spleen, the colloidal carbon uptake was concentrated in the marginal zone around the white pulp, with an inexpressive uptake of pigments by macrophages from white and red pulps. CONCLUSIONS The X-rays in the rat abdomen are associated with a reduction in the Kupffer cells uptake of colloidal carbon, hepatocyte disorders, bile duct proliferation, and splenic uptake of colloidal carbon concentrated in the marginal zone.
Collapse
Affiliation(s)
- Fernando Pereira
- Universidade Federal de Minas Gerais - Departamento de Engenharia Nuclear - Belo Horizonte (MG) - Brazil
| | - Andy Petroianu
- Universidade Federal de Minas Gerais - Departamento de Cirurgia - Belo Horizonte (MG) - Brazil
| | | | - Claubia Pereira
- Universidade Federal de Minas Gerais - Departamento de Engenharia Nuclear - Belo Horizonte (MG) - Brazil
| |
Collapse
|
24
|
Zhang Y, Lv N, Li M, Liu M, Wu C. Cancer-associated fibroblasts: tumor defenders in radiation therapy. Cell Death Dis 2023; 14:541. [PMID: 37607935 PMCID: PMC10444767 DOI: 10.1038/s41419-023-06060-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 05/24/2023] [Accepted: 07/10/2023] [Indexed: 08/24/2023]
Abstract
Cancer-associated fibroblasts (CAFs) are an important component of the tumor microenvironment that are involved in multiple aspects of cancer progression and considered contributors to tumor immune escape. CAFs exhibit a unique radiation resistance phenotype, and can survive clinical radiation doses; however, ionizing radiation can induce changes in their secretions and influence tumor progression by acting on tumor and immune cells. In this review, we describe current knowledge of the effects of radiation therapies on CAFs, as well as summarizing understanding of crosstalk among CAFs, tumor cells, and immune cells. We highlight the important role of CAFs in radiotherapy resistance, and discuss current and future radiotherapy strategies for targeting CAFs.
Collapse
Affiliation(s)
- Yalin Zhang
- Department of Radiation Oncology, Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Na Lv
- Department of Radiation Oncology, Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Manshi Li
- Department of Radiation Oncology, Fourth Affiliated Hospital of China Medical University, Liaoning, China
| | - Ming Liu
- Department of Clinical Epidemiology, Fourth Affiliated Hospital of China Medical University, Liaoning, China.
| | - Chunli Wu
- Department of Radiation Oncology, Fourth Affiliated Hospital of China Medical University, Liaoning, China.
| |
Collapse
|
25
|
Zhao F, Tian H, Wang Y, Zhang J, Liu F, Fu L. LINC01004-SPI1 axis-activated SIGLEC9 in tumor-associated macrophages induces radioresistance and the formation of immunosuppressive tumor microenvironment in esophageal squamous cell carcinoma. Cancer Immunol Immunother 2023; 72:1835-1851. [PMID: 36688997 DOI: 10.1007/s00262-022-03364-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/27/2022] [Indexed: 01/24/2023]
Abstract
Radioresistance and immunosuppression remain the major obstacles in the anti-cancer treatments. This work studies the functions of sialic acid binding Ig like lectin 9 (SIGLEC9) and its related molecules in radioresistance and immunosuppression in esophageal squamous cell carcinoma (ESCC). The single-cell analysis showed that SIGLEC9 was mainly expressed on tumor-associated macrophages (TAMs). Monocytes-derived macrophages were co-cultured with ESCC cells and subjected to radiotherapy. High or low doses of radiotherapy induced SIGLEC9 upregulation and M2 polarization of TAMs. Artificial inhibition of SIGLEC9 in TAMs suppressed the radioresistance and immunosuppressive tumor microenvironment (TME) in the co-cultured ESCC cells. Upstream molecules of SIGLEC9 were predicted via bioinformatics. LINC01004 recruited Spi-1 proto-oncogene (SPI1) in nucleus of TAMs to induce transcriptional activation of SIGLEC9. SIGLEC9 interacted with mucin 1 (MUC1). MUC1 overexpression in ESCCs induced M2 skewing of TAMs, enhanced radioresistance and immunosuppression, and promoted nuclear translocation of β-catenin to suppress radiotherapy-induced ferroptosis of ESCC cells. These effects were blocked upon SIGLEC9 suppression. In vitro results were reproduced in the animal models with xenograft tumors. Taken together, this study demonstrates that the LINC01004-SPI1 axis-activated SIGLEC9 in TAMs induces radioresistance and the formation of immunosuppressive TME in ESCC.
Collapse
Affiliation(s)
- Fen Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, Shandong, People's Republic of China
| | - Hui Tian
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, People's Republic of China
| | - Yungang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Jianbo Zhang
- Departments of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China
| | - Fang Liu
- Department of Imaging, Shandong Medical College, Jinan, 250002, Shandong, People's Republic of China
| | - Lei Fu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
26
|
Deloch L, Rückert M, Weissmann T, Lettmaier S, Titova E, Wolff T, Weinrich F, Fietkau R, Gaipl US. The various functions and phenotypes of macrophages are also reflected in their responses to irradiation: A current overview. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 376:99-120. [PMID: 36997271 DOI: 10.1016/bs.ircmb.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Macrophages are a vital part of the innate immune system that are involved in healthy biological processes but also in disease modulation and response to therapy. Ionizing radiation is commonly used in the treatment of cancer and, in a lower dose range, as additive therapy for inflammatory diseases. In general, lower doses of ionizing radiation are known to induce rather anti-inflammatory responses, while higher doses are utilized in cancer treatment where they result, next to tumor control, in rather inflammatory responses. Most experiments that have been carried out in ex vivo on macrophages find this to be true, however in vivo, tumor-associated macrophages, for example, show a contradictory response to the respective dose-range. While some knowledge in radiation-induced modulations of macrophages has been collected, many of the underlying mechanisms remain unclear. Due to their pivotal role in the human body, however, they are a great target in therapy and could potentially aid in better treatment outcome. We therefore summarized the current knowledge of macrophage mediated radiation responses.
Collapse
|
27
|
Wang NH, Lei Z, Yang HN, Tang Z, Yang MQ, Wang Y, Sui JD, Wu YZ. Radiation-induced PD-L1 expression in tumor and its microenvironment facilitates cancer-immune escape: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1406. [PMID: 36660640 PMCID: PMC9843429 DOI: 10.21037/atm-22-6049] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
Background and Objective Radiotherapy (RT) is one of the fundamental anti-cancer regimens by means of inducing in situ tumor vaccination and driving a systemic anti-tumor immune response. It can affect the tumor microenvironment (TME) components consisting of blood vessels, immunocytes, fibroblasts, and extracellular matrix (ECM), and might subsequently suppress anti-tumor immunity through expression of molecules such as programmed death ligand-1 (PD-L1). Immune checkpoint inhibitors (ICIs), especially anti-programmed cell death 1 (PD-1)/PD-L1 therapies, have been regarded as effective in the reinvigoration of the immune system and another major cancer treatment. Experimentally, combination of RT and ICIs therapy shows a greater synergistic effect than either therapy alone. Methods We performed a narrative review of the literature in the PubMed database. The research string comprised various combinations of "radiotherapy", "programmed death-ligand 1", "microenvironment", "exosome", "myeloid cell", "tumor cell", "tumor immunity". The database was searched independently by two authors. A third reviewer mediated any discordance of the results of the two screeners. Key Content and Findings RT upregulates PD-L1 expression in tumor cells, tumor-derived exosomes (TEXs), myeloid-derived suppressor cells (MDSCs), and macrophages. The signaling pathways correlated to PD-L1 expression in tumor cells include the DNA damage signaling pathway, epidermal growth factor receptor (EGFR) pathway, interferon gamma (IFN-γ) pathway, cGAS-STING pathway, and JAK/STATs pathway. Conclusions PD-L1 upregulation post-RT is found not only in tumor cells but also in the TME and is one of the mechanisms of tumor evasion. Therefore, further studies are necessary to fully comprehend this biological process. Meanwhile, combination of therapies has been shown to be effective, and novel approaches are to be developed as adjuvant to RT and ICIs therapy.
Collapse
Affiliation(s)
- Nuo-Han Wang
- College of Medicine, Chongqing University, Chongqing, China
| | - Zheng Lei
- College of Medicine, Chongqing University, Chongqing, China
| | - Hao-Nan Yang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Zheng Tang
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Meng-Qi Yang
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Ying Wang
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Jiang-Dong Sui
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yong-Zhong Wu
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
28
|
Concerted regulation of OPG/RANKL/ NF‑κB/MMP-13 trajectories contribute to ameliorative capability of prodigiosin and/or low dose γ-radiation against adjuvant- induced arthritis in rats. Int Immunopharmacol 2022; 111:109068. [PMID: 35944459 DOI: 10.1016/j.intimp.2022.109068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prodigiosin (PDG) is a microbial red dye with antioxidant and anti-inflammatory properties, although its effect on rheumatoid arthritis (RA) remains uncertain. Also, multiple doses of low dose γ- radiation (LDR) have been observed to be as a successful intervention for RA. Thus, the purpose of this study was to investigate the ameliorative potential of PDG and/or LDR on adjuvant-induced arthritis (AIA) in rats. METHODS The anti-inflammatory and anti-arthritic effects of PDG and/or LDR were examined in vitro and in vivo, respectively. In the AIA model, the arthritic indexes, paw swelling degrees, body weight gain, and histopathological assessment in AIA rats were assayed. The impact of PDG (200 µg/kg; p.o) and/or LDR (0.5 Gy) on the levels of pro- and anti-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-18, IL-17A, and IL-10) as well as the regulation of osteoprotegrin (OPG)/ receptor activator of nuclear factor κB ligand (RANKL)/ nuclear factor-κB (NF-κB)/MMP-13 pathways was determined. Methotrexate (MTX; 0.05 mg/kg; twice/week, i.p) was administered concurrently as a standard anti-arthritic drug. RESULTS PDG and/or LDR markedly diminished the arthritic indexes, paw edema, weigh loss in AIA rats, alleviated the pathological alterations in joints, reduced the levels of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, IL-18, IL-17A, and RANKL in serum and synovial tissues, while increasing anti-inflammatory cytokines IL-10 and OPG levels. Moreover, PDG and/or LDR down-regulated the expression of RANKL, NF-κBp65, MMP13, caspase-3, and decreased the RANKL/OPG ratio, whereas OPG and collagen II were enhanced in synovial tissues. CONCLUSION PDG and/or LDR exhibited obvious anti-RA activity on AIA.
Collapse
|
29
|
Ozyurek Y, Sari SY, Hurmuz P. Comments on "Whole lung irradiation as a novel treatment for COVID-19: Final results of the prospective randomized trial (WINCOVID trial)". Radiother Oncol 2022; 170:244-245. [PMID: 35259417 PMCID: PMC8897284 DOI: 10.1016/j.radonc.2022.02.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/28/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Yasin Ozyurek
- Department of Radiation Oncology, Hacettepe University, Faculty of Medicine, Ankara, Turkey.
| | - Sezin Yuce Sari
- Department of Radiation Oncology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Pervin Hurmuz
- Department of Radiation Oncology, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
30
|
Dove AP, Cmelak A, Darrow K, McComas KN, Chowdhary M, Beckta J, Kirschner AN. The Use of Low-Dose Radiotherapy in Osteoarthritis: A Review. Int J Radiat Oncol Biol Phys 2022; 114:203-220. [DOI: 10.1016/j.ijrobp.2022.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/08/2022] [Accepted: 04/21/2022] [Indexed: 11/26/2022]
|
31
|
Abstract
PURPOSE Low dose radiation therapy (LDRT) using doses in the range of 30-150 cGy has been proposed as a means of mitigating the pneumonia associated with COVID-19. However, preliminary results from ongoing clinical trials have been mixed. The aim of this work is to develop a mathematical model of the viral infection and associated systemic inflammation in a patient based on the time evolution of the viral load. The model further proposes an immunomodulatory response to LDRT based on available data. Inflammation kinetics are then explored and compared to clinical results. METHODS The time evolution of a viral infection, inflammatory signaling factors, and inflammatory response are modeled by a set of coupled differential equations. Adjustable parameters are taken from the literature where available and otherwise iteratively adjusted to fit relevant data. Simple functions modeling both the suppression of pro-inflammatory signal factors and the enhancement of anti-inflammatory factors in response to low doses of radiation are developed. The inflammation response is benchmarked against C-reactive protein (CRP) levels measured for cohorts of patients with severe COVID-19. RESULTS The model fit the time-evolution of viral load data, cytokine data, and inflammation (CRP) data. When LDRT was applied early, the model predicted a reduction in peak inflammation consistent with the difference between the non-surviving and surviving cohorts. This reduction of peak inflammation diminished as the application of LDRT was delayed. CONCLUSION The model tracks the available data on viral load, cytokine levels, and inflammatory biomarkers well. An LDRT effect is large enough in principle to provide a life-saving immunomodulatory effect, though patients treated with LDRT already near the peak of their inflammation trajectory are unlikely to see drastic reductions in that peak. This result potentially explains some discrepancies in the preliminary clinical trial data.
Collapse
Affiliation(s)
- Charles Kirkby
- Department of Medical Physics, Jack Ady Cancer Centre, Lethbridge, Alberta, Canada.,Department of Oncology, University of Calgary, Calgary, Alberta, Canada.,Department of Physics and Astronomy, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
32
|
Chargari C, Rassy E, Helissey C, Achkar S, Francois S, Deutsch E. Impact of radiation therapy on healthy tissues. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 376:69-98. [PMID: 36997270 DOI: 10.1016/bs.ircmb.2022.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Radiation therapy has a fundamental role in the management of cancers. However, despite a constant improvement in radiotherapy techniques, the issue of radiation-induced side effects remains clinically relevant. Mechanisms of acute toxicity and late fibrosis are therefore important topics for translational research to improve the quality of life of patients treated with ionizing radiations. Tissue changes observed after radiotherapy are consequences of complex pathophysiology, involving macrophage activation, cytokine cascade, fibrotic changes, vascularization disorders, hypoxia, tissue destruction and subsequent chronic wound healing. Moreover, numerous data show the impact of these changes in the irradiated stroma on the oncogenic process, with interplays between tumor radiation response and pathways involved in the fibrotic process. The mechanisms of radiation-induced normal tissue inflammation are reviewed, with a focus on the impact of the inflammatory process on the onset of treatment-related toxicities and the oncogenic process. Possible targets for pharmacomodulation are also discussed.
Collapse
|
33
|
Fang J, Lu J, Zhang Y, Wang J, Wang S, Fan H, Zhang J, Dai W, Gao J, Yu H. Structural properties, antioxidant and immune activities of low molecular weight peptides from soybean dregs (Okara). Food Chem X 2021; 12:100175. [PMID: 34917928 PMCID: PMC8645904 DOI: 10.1016/j.fochx.2021.100175] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/16/2021] [Accepted: 11/29/2021] [Indexed: 01/31/2023] Open
Abstract
In this study, a method for preparing low molecular weight peptides (HPH-VAP) from okara using high-pressure homogenization assisted double enzymes was proposed. In order to explore its advantages, the effects of various methods on protein extraction rate and on the structure, antioxidant and immune properties of peptides were compared. The results showed that the protein extraction rate of this method was increased by 69% and 51% compared with other methods, and the structure only led to changes in the hydrogen bonds between peptide chains. HPH-VAP was screened out through functional characteristics, its structure was identified by HPLC-MS/MS, and further immunological activity analysis was carried out. The results showed that it promoted cell phagocytic ability, NO level and release of cytokines IL-6, IFN- γ, TNF-α. Therefore, this method is an effective and applicable method for industrial preparation of okara peptides, and has a positive effect on the reuse of okara resources.
Collapse
Affiliation(s)
- Jiaqi Fang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Jiahong Lu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Ying Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Jinyu Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Hongliang Fan
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Jiarui Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Weichang Dai
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| | - Junpeng Gao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China.,National Soybean Industry Technology System Processing Laboratory, Jilin, Changchun 130118, China
| |
Collapse
|
34
|
Abdel-Rafei MK, Thabet NM, El Tawel G, El Bakary NM, El Fatih NM, Sh Azab K. Role of leptin/STAT3 signaling and RIP-kinases in fucoxanthin influences on mice exposed to LPS and gamma radiation. TOXIN REV 2021. [DOI: 10.1080/15569543.2021.2008451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Mohamed K. Abdel-Rafei
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Noura M. Thabet
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Ghada El Tawel
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Nermeen M. El Bakary
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Neama M. El Fatih
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Khaled Sh Azab
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
35
|
Donaubauer AJ, Becker I, Weissmann T, Fröhlich BM, Muñoz LE, Gryc T, Denzler M, Ott OJ, Fietkau R, Gaipl US, Frey B. Low Dose Radiation Therapy Induces Long-Lasting Reduction of Pain and Immune Modulations in the Peripheral Blood - Interim Analysis of the IMMO-LDRT01 Trial. Front Immunol 2021; 12:740742. [PMID: 34712229 PMCID: PMC8546320 DOI: 10.3389/fimmu.2021.740742] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
The treatment of chronic inflammatory and degenerative diseases by low dose radiation therapy (LDRT) is promising especially for patients who were refractory for classical therapies. LDRT aims to reduce pain of patients and to increase their mobility. Although LDRT has been applied since the late 19th century, the immunological mechanisms remain elusive. Within the prospective IMMO-LDRT01 trial (NCT02653079) the effects of LDRT on the peripheral blood immune status, as well as on pain and life quality of patients have been analyzed. Blood is taken before and after every serial irradiation with a single dose per fraction of 0.5Gy, as well as during follow-up appointments in order to determine a detailed longitudinal immune status by multicolor flow cytometry. Here, we report the results of an interim analysis of 125 patients, representing half the number of patients to be recruited. LDRT significantly improved patients’ pain levels and induced distinct systemic immune modulations. While the total number of leukocytes remained unchanged in the peripheral blood, LDRT induced a slight reduction of eosinophils, basophils and plasmacytoid dendritic cells and an increase of B cells. Furthermore, activated immune cells were decreased following LDRT. Especially cells of the monocytic lineage correlated to LDRT-induced improvements of clinical symptoms, qualifying these immune cells as predictive biomarkers for the therapeutic success. We conclude that LDRT improves pain of the patients by inducing systemic immune modulations and that immune biomarkers could be defined for prediction by improved patient stratification in the future.
Collapse
Affiliation(s)
- Anna-Jasmina Donaubauer
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Ina Becker
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Thomas Weissmann
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Birgitta M Fröhlich
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Luis E Muñoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, Germany
| | - Thomas Gryc
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Manuel Denzler
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Oliver J Ott
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
36
|
Wen L, Jiang Y, Zhou X, Bi H, Yang B. Structure identification of soybean peptides and their immunomodulatory activity. Food Chem 2021; 359:129970. [PMID: 34015561 DOI: 10.1016/j.foodchem.2021.129970] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/15/2021] [Accepted: 04/25/2021] [Indexed: 10/21/2022]
Abstract
Soybean peptides are functional food with good health benefits. The health benefits presented are highly dependent on the peptide structure. In this work, soybean peptides were prepared by alkaline protease hydrolysis of soybean proteins. The peptide structure was identified by UPLC-MS/MS. The full peptide composition was revealed. The sequences of 51 peptides were identified and 46 peptides were assigned as immunomodulatory peptides. By evaluating the immumonodulatory activity and mechanism, soybean peptides could facilitate the proliferation of macrophages. The pinocytotic activity and NO level were increased. Induction of iNOS mRNA expression by soybean peptides was responsible for the increased NO production. The release of cytokines IL-6 and TNF-α was elevated and their levels were equal to positive control. The mRNA expression levels of IL-6 and TNF-α were also improved by soybean peptides, but much lower than positive control. The results were helpful for application of soybean peptides in functional foods.
Collapse
Affiliation(s)
- Lingrong Wen
- Guangdong Provincial Key Laboratory of Applied Botany, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Yueming Jiang
- Guangdong Provincial Key Laboratory of Applied Botany, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China
| | - Xuesong Zhou
- Guangzhou Honsea Industry Co., Ltd., Guangzhou 510530, China
| | - Huimin Bi
- Guangzhou College of Technology and Business, Guangzhou 510850, China
| | - Bao Yang
- Guangdong Provincial Key Laboratory of Applied Botany, Key Laboratory of Post-Harvest Handling of Fruits, Ministry of Agriculture, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China.
| |
Collapse
|
37
|
Javadinia SA, Nazeminezhad N, Ghahramani-Asl R, Soroosh D, Fazilat-Panah D, PeyroShabany B, Saberhosseini SN, Mehrabian A, Taghizadeh-Hesary F, Nematshahi M, Dhawan G, Welsh JS, Calabrese EJ, Kapoor R. Low-dose radiation therapy for osteoarthritis and enthesopathies: a review of current data. Int J Radiat Biol 2021; 97:1352-1367. [PMID: 34259615 DOI: 10.1080/09553002.2021.1956000] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Osteoarthritis (OA), the most common degenerative joint disease, is associated with severe functional limitation and impairment of quality of life. Numerous reports have documented the clinical efficacy of low-dose radiotherapy (LD-RT) in the management of various inflammatory disorders, including OA. In this paper, we assessed the clinical literature involving the use of LD-RT in the treatment of OA, its dose-response features, possible underlying mechanistic features, and optimal therapeutic dose range. METHODS We carried out a systematic review based on the guidelines of the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) statements and evaluated articles meeting the inclusion criteria for this review. RESULTS A total of 361 articles were identified from databases, such as Scopus, PubMed, Embase, and Science Direct out of which 224 articles were duplicates and were discarded. Of the remaining 137 articles, 74 articles were un-related, 27 articles were review articles, eight were conference abstracts, three were letters, two were editorials, two were notes, and one was a book chapter. Finally, 20 articles met all the inclusion criteria and were included in this systematic review. DISCUSSION Several single-arm retrospective/prospective studies showed advantages for LD-RT in the management of OA in terms of pain relief, improvement of mobility and function, and showed minimal side effects. Mechanistic considerations involve positive subcellular effects mediated by the activation of a nuclear factor erythroid 2-related transcription factor (Nrf2) mediated antioxidant response. Further research on both the short- and long-term effects of LD-RT on OA and other inflammatory disorders is recommended.
Collapse
Affiliation(s)
- Seyed Alireza Javadinia
- Clinical Research Development Unit, Hospital Research Development Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | | - Ruhollah Ghahramani-Asl
- Clinical Research Development Unit, Hospital Research Development Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Soroosh
- Clinical Research Development Unit, Hospital Research Development Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | | - Babak PeyroShabany
- Department of Internal Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | | - Arezoo Mehrabian
- Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mohammad Nematshahi
- Department of Anesthesiology and Critical Care, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Gaurav Dhawan
- Sri Guru Ram Das University of Health Sciences, Amritsar, India
| | - James S Welsh
- Edward Hines Jr. VA Hospital, Loyola University Chicago Stritch School of Medicine, Chicago, IL, USA
| | - Edward J Calabrese
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, USA
| | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, CT, USA
| |
Collapse
|
38
|
Khan AUH, Blimkie M, Yang DS, Serran M, Pack T, Wu J, Kang JY, Laakso H, Lee SH, Le Y. Effects of Chronic Low-Dose Internal Radiation on Immune-Stimulatory Responses in Mice. Int J Mol Sci 2021; 22:7303. [PMID: 34298925 PMCID: PMC8306076 DOI: 10.3390/ijms22147303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 12/18/2022] Open
Abstract
The Linear-No-Threshold (LNT) model predicts a dose-dependent linear increase in cancer risk. This has been supported by biological and epidemiological studies at high-dose exposures. However, at low-doses (LDR ≤ 0.1 Gy), the effects are more elusive and demonstrate a deviation from linearity. In this study, the effects of LDR on the development and progression of mammary cancer in FVB/N-Tg(MMTVneu)202Mul/J mice were investigated. Animals were chronically exposed to total doses of 10, 100, and 2000 mGy via tritiated drinking water, and were assessed at 3.5, 6, and 8 months of age. Results indicated an increased proportion of NK cells in various organs of LDR exposed mice. LDR significantly influenced NK and T cell function and activation, despite diminishing cell proliferation. Notably, the expression of NKG2D receptor on NK cells was dramatically reduced at 3.5 months but was upregulated at later time-points, while the expression of NKG2D ligand followed the opposite trend, with an increase at 3.5 months and a decrease thereafter. No noticeable impact was observed on mammary cancer development, as measured by tumor load. Our results demonstrated that LDR significantly influenced the proportion, proliferation, activation, and function of immune cells. Importantly, to the best of our knowledge, this is the first report demonstrating that LDR modulates the cross-talk between the NKG2D receptor and its ligands.
Collapse
Affiliation(s)
- Abrar Ul Haq Khan
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Melinda Blimkie
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Doo Seok Yang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Mandy Serran
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Tyler Pack
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Jin Wu
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Ji-Young Kang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
| | - Holly Laakso
- Radiobiology and Health Branch, Canadian Nuclear Laboratories Ltd., Chalk River, ON K0J 1J0, Canada; (M.B.); (M.S.); (T.P.); (J.W.); (H.L.)
| | - Seung-Hwan Lee
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
- Centre for Infection, The University of Ottawa, Immunity and Inflammation, Ottawa, ON K1H 8M5, Canada
| | - Yevgeniya Le
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (A.U.H.K.); (D.S.Y.); (J.-Y.K.)
- CANDU Owners Group Inc., Toronto, ON M5G 2K4, Canada
| |
Collapse
|
39
|
Calabrese EJ, Kozumbo WJ, Kapoor R, Dhawan G, Lara PC, Giordano J. Nrf2 activation putatively mediates clinical benefits of low-dose radiotherapy in COVID-19 pneumonia and acute respiratory distress syndrome (ARDS): Novel mechanistic considerations. Radiother Oncol 2021; 160:125-131. [PMID: 33932453 PMCID: PMC8080499 DOI: 10.1016/j.radonc.2021.04.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022]
Abstract
Novel mechanistic insights are discussed herein that link a single, nontoxic, low-dose radiotherapy (LDRT) treatment (0.5-1.0 Gy) to (1) beneficial subcellular effects mediated by the activation of nuclear factor erythroid 2-related transcription factor (Nrf2) and to (2) favorable clinical outcomes for COVID-19 pneumonia patients displaying symptoms of acute respiratory distress syndrome (ARDS). We posit that the favorable clinical outcomes following LDRT result from potent Nrf2-mediated antioxidant responses that rebalance the oxidatively skewed redox states of immunological cells, driving them toward anti-inflammatory phenotypes. Activation of Nrf2 by ionizing radiation is highly dose dependent and conforms to the features of a biphasic (hormetic) dose-response. At the cellular and subcellular levels, hormetic doses of <1.0 Gy induce polarization shifts in the predominant population of lung macrophages, from an M1 pro-inflammatory to an M2 anti-inflammatory phenotype. Together, the Nrf2-mediated antioxidant responses and the subsequent shifts to anti-inflammatory phenotypes have the capacity to suppress cytokine storms, resolve inflammation, promote tissue repair, and prevent COVID-19-related mortality. Given these mechanistic considerations-and the historical clinical success of LDRT early in the 20th century-we opine that LDRT should be regarded as safe and effective for use at almost any stage of COVID-19 infection. In theory, however, optimal life-saving potential is thought to occur when LDRT is applied prior to the cytokine storms and before the patients are placed on mechanical oxygen ventilators. The administration of LDRT either as an intervention of last resort or too early in the disease progression may be far less effective in saving the lives of ARDS patients.
Collapse
Affiliation(s)
| | | | - Rachna Kapoor
- Saint Francis Hospital and Medical Center, Hartford, USA
| | - Gaurav Dhawan
- Sri Guru Ram Das University of Health Sciences, Amritsar, India.
| | - Pedro C Lara
- Department of Radiation Oncology, Hospital Universitario San Roque, Universidad Fernando Pessoa Canarias, Las Palmas Gran Canaria, Spain.
| | - James Giordano
- Departments of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC, USA.
| |
Collapse
|
40
|
Winn BJ, Kersten RC. Teprotumumab: Interpreting the Clinical Trials in the Context of Thyroid Eye Disease Pathogenesis and Current Therapies. Ophthalmology 2021; 128:1627-1651. [PMID: 33930408 DOI: 10.1016/j.ophtha.2021.04.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 11/16/2022] Open
Abstract
Teprotumumab, a monoclonal antibody targeted against the insulin-like growth factor 1 (IGF-1) receptor, was recently approved by the United States Food and Drug Administration for the treatment of thyroid eye disease (TED). Phase 1 studies of teprotumumab for the treatment of malignancies demonstrated an acceptable safety profile but limited effectiveness. Basic research implicating the IGF-1 receptor on the CD-34+ orbital fibrocyte in the pathogenesis of TED renewed interest in the drug. Two multicenter, randomized, double-masked, clinical trials (phase 2 and 3) evaluated the efficacy of 8 infusions of teprotumumab every 3 weeks versus placebo in 170 patients with recent-onset active TED, as defined by a clinical activity score (CAS) of at least 4. Teprotumumab was superior to placebo for the primary efficacy end points in both studies: overall responder rate as defined by a reduction of 2 or more CAS points and a reduction of 2 mm or more in proptosis (69% vs. 20%; P < 0.001; phase 2 study) and proptosis responder rate as defined by a reduction of 2 mm or more in proptosis (83% vs. 10%; P < 0.001; phase 3 study). In both studies, treatment with teprotumumab compared with placebo achieved a significant mean reduction of proptosis (-3.0 mm vs. -0.3 mm, phase 2 study; -3.32 mm vs. -0.53 mm, phase 3 study) and CAS (-4.0 vs. -2.5, phase 2 study; -3.7 vs. -2.0, phase 3 study). Teprotumumab also resulted in a greater proportion of patients with a final CAS of 0 or 1, higher diplopia responder rate, and a larger improvement in the Graves' Ophthalmopathy Quality of Life overall score. More than half of patients (62%, phase 2 trial; 56%, phase 3 trial) who were primary end point responders maintained this response at 51 weeks after the last dose of therapy. The most common adverse events reported with teprotumumab included muscle spasms (25%), nausea (17%), alopecia (13%), diarrhea (13%), fatigue (10%), hearing impairment (10%), and hyperglycemia (8%). Teprotumumab is contraindicated for those with inflammatory bowel disease and who are pregnant. Although the current dosing regimen has proven effective for TED, dose-ranging studies including variable concentrations, infusion frequencies, and durations of teprotumumab therapy in the setting of TED have not been performed.
Collapse
Affiliation(s)
- Bryan J Winn
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California; Ophthalmology Section, Surgical Service, San Francisco Veterans Affairs Medical Center, San Francisco, California.
| | - Robert C Kersten
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
41
|
Low- vs. high-dose radiotherapy in Graves' ophthalmopathy: a retrospective comparison of long-term results. Strahlenther Onkol 2021; 197:885-894. [PMID: 33860819 PMCID: PMC8458186 DOI: 10.1007/s00066-021-01770-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/16/2021] [Indexed: 11/22/2022]
Abstract
Purpose Radiotherapy represents an effective treatment option in Graves’ ophthalmopathy (GO), leading to palliation of clinical symptoms. However, there are only a limited number of trials comparing the effectiveness of low- vs. high-dose radiotherapy. Methods We analyzed 127 patients treated with radiotherapy for stage 3/4 GO (NOSPECS classification). Patients were treated with single doses of 2.0 Gy (cumulative dose 20 Gy) until 2007, afterwards a single dose of 0.8 Gy (cumulative dose 4.8 Gy) was applied. With a median follow-up-time of 9.0 years, the treatment efficacy (overall improvement, sense of eye pressure, lid edema, ocular motility, exophthalmos, subjective vision, and diplopia) and adverse effects were analyzed by a standardized survey. Results Overall, 63.8% described improvement of symptoms after radiotherapy. No significant differences in overall treatment response and improvement of main outcome measures between low- or high-dose radiotherapy treatments are detectable, while low-dose radiotherapy leads significantly more often to retreatment (13.1% vs. 1.7%, p = 0.016). The main independent predictor of treatment response is the presence of lid edema (odds ratio, OR, 3.53; p = 0.006). Conclusion At long-term follow-up, the majority of patients reported palliation of symptoms with limited adverse effects, suggesting clinical effectiveness of radiotherapy for amelioration of GO symptoms independent of low- or high-dose radiotherapy.
Collapse
|
42
|
Lumniczky K, Impens N, Armengol G, Candéias S, Georgakilas AG, Hornhardt S, Martin OA, Rödel F, Schaue D. Low dose ionizing radiation effects on the immune system. ENVIRONMENT INTERNATIONAL 2021; 149:106212. [PMID: 33293042 PMCID: PMC8784945 DOI: 10.1016/j.envint.2020.106212] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/20/2020] [Accepted: 09/03/2020] [Indexed: 05/03/2023]
Abstract
Ionizing radiation interacts with the immune system in many ways with a multiplicity that mirrors the complexity of the immune system itself: namely the need to maintain a delicate balance between different compartments, cells and soluble factors that work collectively to protect, maintain, and restore tissue function in the face of severe challenges including radiation damage. The cytotoxic effects of high dose radiation are less relevant after low dose exposure, where subtle quantitative and functional effects predominate that may go unnoticed until late after exposure or after a second challenge reveals or exacerbates the effects. For example, low doses may permanently alter immune fitness and therefore accelerate immune senescence and pave the way for a wide spectrum of possible pathophysiological events, including early-onset of age-related degenerative disorders and cancer. By contrast, the so called low dose radiation therapy displays beneficial, anti-inflammatory and pain relieving properties in chronic inflammatory and degenerative diseases. In this review, epidemiological, clinical and experimental data regarding the effects of low-dose radiation on the homeostasis and functional integrity of immune cells will be discussed, as will be the role of immune-mediated mechanisms in the systemic manifestation of localized exposures such as inflammatory reactions. The central conclusion is that ionizing radiation fundamentally and durably reshapes the immune system. Further, the importance of discovery of immunological pathways for modifying radiation resilience amongst other research directions in this field is implied.
Collapse
Affiliation(s)
- Katalin Lumniczky
- National Public Health Centre, Department of Radiation Medicine, Budapest, Albert Florian u. 2-6, 1097, Hungary.
| | - Nathalie Impens
- Belgian Nuclear Research Centre, Biosciences Expert Group, Boeretang 200, 2400 Mol, Belgium.
| | - Gemma Armengol
- Unit of Biological Anthropology, Department of Animal Biology, Plant Biology and Ecology, Faculty of Biosciences, Universitat Autònoma de Barcelona, 08193-Bellaterra, Barcelona, Catalonia, Spain.
| | - Serge Candéias
- Université Grenoble-Alpes, CEA, CNRS, IRIG-LCBM, 38000 Grenoble, France.
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou 15780, Athens, Greece.
| | - Sabine Hornhardt
- Federal Office for Radiation Protection (BfS), Ingolstaedter Landstr.1, 85764 Oberschleissheim, Germany.
| | - Olga A Martin
- Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne 3052, Victoria, Australia.
| | - Franz Rödel
- Department of Radiotherapy and Oncology, University Hospital, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA 90095-1714, USA.
| |
Collapse
|
43
|
Ameri A, Ameri P, Rahnama N, Mokhtari M, Sedaghat M, Hadavand F, Bozorgmehr R, Haghighi M, Taghizadeh-Hesary F. Low-Dose Whole-Lung Irradiation for COVID-19 Pneumonia: Final Results of a Pilot Study. Int J Radiat Oncol Biol Phys 2021; 109:859-866. [PMID: 33278503 PMCID: PMC7709599 DOI: 10.1016/j.ijrobp.2020.11.065] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/21/2020] [Accepted: 11/26/2020] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Radiation therapy (RT), commonly used in cancer management, has been considered as one of the potential treatments for COVID-19 pneumonia. Here, we present the results of the pilot trial evaluating low-dose whole-lung irradiation (LD-WLI) in patients with COVID-19 pneumonia. METHODS Ten patients with moderate COVID-19 pneumonia were treated with LD-WLI in a single fraction of 0.5 or 1.0 Gy along with the national protocol. The primary endpoint was an improvement in Spo2. The secondary endpoints were the number of days of hospital/intensive care unit stay, the number of intubations after RT, 28-day mortality, and changes in biomarkers. The response rate (RR) was defined as an increase in Spo2 upon RT with a rising or constant trend in the next 2 days, clinical recovery (CR) including patients who were discharged or acquired Spo2 ≥93% on room air, and 28-day mortality rate defined based on days of RT. RESULTS The median age was 75 years (80% male). Five, 1, and 4 patients received single-dose 0.5 Gy, two-dose 0.5 Gy, and single-dose 1.0 Gy LD-WLI, respectively. The mean improvement in Spo2 at days 1 and 2 after RT was 2.4% (±4.8%) and 3.6% (±6.1%), respectively, with improvement in 9 patients after 1 day. Five, 1, and 4 patients were discharged, opted out of the trial, and died in the hospital, respectively. Two of 5 discharged patients died within 3 days at home. Among discharged patients, the Spo2 at discharge was 81% to 88% in 3 patients and 93% in the other 2 patients. Overall, the RR and CR were 63.6% and 55.5%, respectively. The RR, CR, and 28-day mortality of the single 0.5 Gy and 1.0 Gy WLI groups were 71.4% versus 50% (P = .57), 60% versus 50% (P = .64), and 50% versus 75% (P = .57), respectively. CONCLUSION LD-WLI with a single fraction of 0.5 Gy or 1 Gy is feasible. A randomized trial with patients who do not receive radiation is required to assess the efficacy of LD-WLI for COVID-19.
Collapse
Affiliation(s)
- Ahmad Ameri
- Department of Clinical Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pooya Ameri
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazanin Rahnama
- Department of Clinical Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Majid Mokhtari
- Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meghdad Sedaghat
- Department of Internal Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Hadavand
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rama Bozorgmehr
- Clinical Research Development Unit, Shohadaye Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Haghighi
- Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
44
|
Tubin S, Khan MK, Gupta S, Jeremic B. Biology of NSCLC: Interplay between Cancer Cells, Radiation and Tumor Immune Microenvironment. Cancers (Basel) 2021; 13:775. [PMID: 33673332 PMCID: PMC7918834 DOI: 10.3390/cancers13040775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/12/2022] Open
Abstract
The overall prognosis and survival of non-small cell lung cancer (NSCLC) patients remain poor. The immune system plays an integral role in driving tumor control, tumor progression, and overall survival of NSCLC patients. While the tumor cells possess many ways to escape the immune system, conventional radiotherapy (RT) approaches, which are directly cytotoxic to tumors, can further add additional immune suppression to the tumor microenvironment by destroying many of the lymphocytes that circulate within the irradiated tumor environment. Thus, the current immunogenic balance, determined by the tumor- and radiation-inhibitory effects is significantly shifted towards immunosuppression, leading to poor clinical outcomes. However, newer emerging evidence suggests that tumor immunosuppression is an "elastic process" that can be manipulated and converted back into an immunostimulant environment that can actually improve patient outcome. In this review we will discuss the natural immunosuppressive effects of NSCLC cells and conventional RT approaches, and then shift the focus on immunomodulation through novel, emerging immuno- and RT approaches that promise to generate immunostimulatory effects to enhance tumor control and patient outcome. We further describe some of the mechanisms by which these newer approaches are thought to be working and set the stage for future trials and additional preclinical work.
Collapse
Affiliation(s)
- Slavisa Tubin
- MedAustron Ion Therapy Center, Marie Curie-Straße 5, 2700 Wiener Neustadt, Austria
| | - Mohammad K. Khan
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA;
| | - Seema Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Branislav Jeremic
- Research Institute of Clinical Medicine, 13 Tevdore Mgdveli, Tbilisi 0112, Georgia;
| |
Collapse
|
45
|
Verigos KE, Sagredou S, Orfanakos K, Dalezis P, Trafalis DT. 8-Hydroxy-2'-Deoxyguanosine and 8-Nitroguanine Production and Detection in Blood Serum of Breast Cancer Patients in Response to Postoperative Complementary External Ionizing Irradiation of Normal Tissues. Dose Response 2021; 18:1559325820982172. [PMID: 33424517 PMCID: PMC7758665 DOI: 10.1177/1559325820982172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022] Open
Abstract
It is widely known that ionizing irradiation is strongly linked to the production of reactive oxygen (ROS) and nitrative species (RNS) through which DNA damage products like 8-hydroxy-2-deoxyguanosine (8-OHdG) and 8-nitroguanine (8-NG) are generated, respectively. In the present study, we aimed to investigate the formation of 8-OHdG and 8-NG upon irradiation and to further explore whether alterations in their concentration levels are related to the administered radiation doses and exposure time. Our research work was conducted in blood serum samples collected from 33 breast cancer patients who received adjuvant radiotherapy. The detection of 8-OHdG and 8-NG was assessed by enzyme-linked immunosorbent assay. Our results suggest that both, 8-OHdG and 8-NG, were formed during the radiation regimen. Significant correlations with radiation dose were also demonstrated by the dose-response curves of 8-OHdG and 8-NG, fitted by logarithmic distribution and polynomial regression, respectively. More precisely, 8-OHdG and 8-NG concentrations (ng/mL) were considerably increased when patients received ionizing radiation up to 30 Gy whereas irradiation over 30 Gy did not induce any further increases. The current study supports a) the production of 8-OHdG and 8-NG during radiotherapy and b) significant correlations between either 8-OHdG or 8-NG levels and radiation doses, indicating a radiation-dose-dependent relationship.
Collapse
Affiliation(s)
- Kosmas E Verigos
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Department of Radiation Therapy, 401 General Military Hospital, Athens, Greece
| | - Sofia Sagredou
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kyriakos Orfanakos
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Radiation Therapy Clinic-A, "Metaxa" Cancer Hospital, Piraeus, Greece
| | - Panayiotis Dalezis
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
46
|
Sayed HM, Said MM, Morcos NYS, El Gawish MA, Ismail AFM. Antitumor and Radiosensitizing Effects of Zinc Oxide-Caffeic Acid Nanoparticles against Solid Ehrlich Carcinoma in Female Mice. Integr Cancer Ther 2021; 20:15347354211021920. [PMID: 34105411 PMCID: PMC8193661 DOI: 10.1177/15347354211021920] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/19/2021] [Accepted: 05/14/2021] [Indexed: 12/24/2022] Open
Abstract
This study aimed to evaluate the anticancer and radio-sensitizing efficacy of Zinc Oxide-Caffeic Acid Nanoparticles (ZnO-CA NPs). ZnO-CA NPs were formulated by the conjugation of Zinc Oxide nanoparticles (ZnO NPs) with caffeic acid (CA) that were characterized by Fourier Transform Infrared Spectra (FT-IR), X-ray Diffractometer (XRD), and Transmission Electron Microscopy (TEM). In vitro anticancer potential of ZnO-CA NPs was evaluated by assessing cell viability in the human breast (MCF-7) and hepatocellular (HepG2) carcinoma cell lines. In vivo anticancer and radio-sensitizing effects of ZnO-CA NPs in solid Ehrlich carcinoma-bearing mice (EC mice) were also assessed. Treatment of EC mice with ZnO-CA NPs resulted in a considerable decline in tumor size and weight, down-regulation of B-cell lymphoma 2 (BCL2) and nuclear factor kappa B (NF-κB) gene expressions, decreased vascular cell adhesion molecule 1 (VCAM-1) level, downregulation of phosphorylated-extracellular-regulated kinase 1 and 2 (p-ERK1/2) protein expression, DNA fragmentation and a recognizable peak at sub-G0/G1 indicating dead cells' population in cancer tissues. Combined treatment of ZnO-CA NPs with γ-irradiation improved these effects. In conclusion: ZnO-CA NPs exhibit in-vitro as well as in-vivo antitumor activity, which is augmented by exposure of mice to γ-irradiation. Further explorations are warranted previous to clinical application of ZnO-CA NPs.
Collapse
Affiliation(s)
- Hayam M. Sayed
- Radiation Biology Department, National
Center for Radiation Research and Technology, Egyptian Atomic Energy Authority,
Cairo, Egypt
| | - Mahmoud M. Said
- Biochemistry Department, Faculty of
Science, Ain Shams University, Cairo, Egypt
| | - Nadia Y. S. Morcos
- Biochemistry Department, Faculty of
Science, Ain Shams University, Cairo, Egypt
| | - Mona A. El Gawish
- Radiation Biology Department, National
Center for Radiation Research and Technology, Egyptian Atomic Energy Authority,
Cairo, Egypt
| | - Amel F. M. Ismail
- Drug Radiation Research Department,
National Center for Radiation Research and Technology, Egyptian Atomic Energy
Authority, Cairo, Egypt
| |
Collapse
|
47
|
Tremi I, Nowsheen S, Aziz K, Siva S, Ventura J, Hatzi VI, Martin OA, Georgakilas AG. Inflammation and oxidatively induced DNA damage: A synergy leading to cancer development. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00013-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
48
|
Obrador E, Salvador R, Villaescusa JI, Soriano JM, Estrela JM, Montoro A. Radioprotection and Radiomitigation: From the Bench to Clinical Practice. Biomedicines 2020; 8:E461. [PMID: 33142986 PMCID: PMC7692399 DOI: 10.3390/biomedicines8110461] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
The development of protective agents against harmful radiations has been a subject of investigation for decades. However, effective (ideal) radioprotectors and radiomitigators remain an unsolved problem. Because ionizing radiation-induced cellular damage is primarily attributed to free radicals, radical scavengers are promising as potential radioprotectors. Early development of such agents focused on thiol synthetic compounds, e.g., amifostine (2-(3-aminopropylamino) ethylsulfanylphosphonic acid), approved as a radioprotector by the Food and Drug Administration (FDA, USA) but for limited clinical indications and not for nonclinical uses. To date, no new chemical entity has been approved by the FDA as a radiation countermeasure for acute radiation syndrome (ARS). All FDA-approved radiation countermeasures (filgrastim, a recombinant DNA form of the naturally occurring granulocyte colony-stimulating factor, G-CSF; pegfilgrastim, a PEGylated form of the recombinant human G-CSF; sargramostim, a recombinant granulocyte macrophage colony-stimulating factor, GM-CSF) are classified as radiomitigators. No radioprotector that can be administered prior to exposure has been approved for ARS. This differentiates radioprotectors (reduce direct damage caused by radiation) and radiomitigators (minimize toxicity even after radiation has been delivered). Molecules under development with the aim of reaching clinical practice and other nonclinical applications are discussed. Assays to evaluate the biological effects of ionizing radiations are also analyzed.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Rosario Salvador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - José M. Soriano
- Food & Health Lab, Institute of Materials Science, University of Valencia, 46980 Valencia, Spain;
- Joint Research Unit in Endocrinology, Nutrition and Clinical Dietetics, University of Valencia-Health Research Institute IISLaFe, 46026 Valencia, Spain
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (E.O.); (R.S.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain;
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
49
|
The Influence of Radiation on Bone and Bone Cells-Differential Effects on Osteoclasts and Osteoblasts. Int J Mol Sci 2020; 21:ijms21176377. [PMID: 32887421 PMCID: PMC7504528 DOI: 10.3390/ijms21176377] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
The bone is a complex organ that is dependent on a tight regulation between bone formation by osteoblasts (OBs) and bone resorption by osteoclasts (OCs). These processes can be influenced by environmental factors such as ionizing radiation (IR). In cancer therapy, IR is applied in high doses, leading to detrimental effects on bone, whereas radiation therapy with low doses of IR is applied for chronic degenerative and inflammatory diseases, with a positive impact especially on bone homeostasis. Moreover, the effects of IR are of particular interest in space travel, as astronauts suffer from bone loss due to space radiation and microgravity. This review summarizes the current state of knowledge on the effects of IR on bone with a special focus on the influence on OCs and OBs, as these cells are essential in bone remodeling. In addition, the influence of IR on the bone microenvironment is discussed. In summary, the effects of IR on bone and bone remodeling cells strongly depend on the applied radiation dose, as differential results are provided from in vivo as well as in vitro studies with varying doses of IR. Furthermore, the isolated effects of IR on a single cell type are difficult to determine, as the bone cells and bone microenvironment are building a tightly regulated network, influencing on one another. Therefore, future research is necessary in order to elucidate the influence of different bone cells on the overall radiation-induced effects on bone.
Collapse
|
50
|
Malekghasemi S, Majidi J, Baghbanzadeh A, Abdolalizadeh J, Baradaran B, Aghebati-Maleki L. Tumor-Associated Macrophages: Protumoral Macrophages in Inflammatory Tumor Microenvironment. Adv Pharm Bull 2020; 10:556-565. [PMID: 33062602 PMCID: PMC7539304 DOI: 10.34172/apb.2020.066] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 02/01/2020] [Accepted: 02/02/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor microenvironment consists of malignant and non-malignant cells. The interaction of these dynamic and different cells is responsible for tumor progression at different levels. The non-malignant cells in TME contain cells such as tumor-associated macrophages (TAMs), cancer associated fibroblasts, pericytes, adipocytes, T cells, B cells, myeloid-derived suppressor cells (MDSCs), tumor-associated neutrophils (TANs), dendritic cells (DCs) and Vascular endothelial cells. TAMs are abundant in most human and murine cancers and their presence are associated with poor prognosis. The major event in tumor microenvironment is macrophage polarization into tumor-suppressive M1 or tumor-promoting M2 types. Although much evidence suggests that TAMS are primarily M2-like macrophages, the mechanism responsible for polarization into M1 and M2 macrophages remain unclear. TAM contributes cancer cell motility, invasion, metastases and angiogenesis. The relationship between TAM and tumor cells lead to used them as a diagnostic marker, therapeutic target and prognosis of cancer. This review presents the origin, polarization, role of TAMs in inflammation, metastasis, immune evasion and angiogenesis as well as they can be used as therapeutic target in variety of cancer cells. It is obvious that additional substantial and preclinical research is needed to support the effectiveness and applicability of this new and promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Somaiyeh Malekghasemi
- Department of Basic Oncology, Oncology Institute, Hacettepe University, Sihhiye, Ankara, TR-06100, Turkey
| | - Jafar Majidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Department of Basic Oncology, Oncology Institute, Hacettepe University, Sihhiye, Ankara, TR-06100, Turkey
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leili Aghebati-Maleki
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|