1
|
Li X, Ding Z. Cognitive dysfunction induced by cranial radiotherapy: mechanisms and therapeutic methods. Brain Res Bull 2024; 218:111106. [PMID: 39447765 DOI: 10.1016/j.brainresbull.2024.111106] [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: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cranial radiotherapy can damage normal brain tissues, inducing cognitive dysfunction in patients. Radiotherapy-induced cognitive dysfunction is associated with hippocampal injury, white matter damage and microvascular injury. In this study, the mechanisms of cognitive dysfunction induced by cranial radiotherapy and combined chemoradiotherapy are reviewed, and the advances in therapeutic methods for radiotherapy-induced brain injury are summarized. The mechanisms of radiotherapy-induced brain injury include a decline of neurogenesis, impairment of neurons and glial cells, vascular injury, oxidative stress and DNA damage, cell death, and inflammatory response. Disruption of the bloodbrain barrier (BBB) increases the exposure of the brain to chemotherapeutic agents, thus exacerbating radiotherapy-induced brain damage. The current methods used to prevent radiotherapy-induced brain injury mainly include precision radiotherapy, stem cell transplantation, and treatment with neuroprotective drugs. The combined application of precision radiotherapy and neuroprotective drugs, including antioxidants, anti-inflammatory agents and other drugs, might exert better neuroprotective effects. To resolve the issues of neuroprotective drugs, such as difficulty in crossing the BBB, nanoenzymes and drug delivery nano-systems could be applied in the future.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
2
|
Pham TN, Coupey J, Ivanova V, Thariat J, Valable S. Differential plasma cytokine variation following X-ray or proton brain irradiation using machine-learning approaches. Cancer Radiother 2024; 28:474-483. [PMID: 39307604 DOI: 10.1016/j.canrad.2024.08.001] [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: 03/29/2024] [Accepted: 08/19/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE X-ray and proton irradiation have been reported to induce distinct modifications in cytokine expression in vitro and in vivo, suggesting a dissimilar inflammatory response between X-rays and protons. We aimed to investigate the differences in cytokine profiles early following fractionated brain irradiation with X-rays or protons and their relationship with leukocyte subpopulations in rodents. MATERIALS AND METHODS Our study utilized data from 80 tumor-free mice subjected to X-ray or proton brain irradiation in four fractions of 2.5Gy. Sixteen non-irradiated mice were used as the controls. Blood was collected 12h postirradiation to examine the profile of 13 cytokines. Correlation analysis, principal component analysis (PCA), and tree-based modeling were used to investigate the relationship between cytokine levels and leukocyte subpopulation variations following irradiation in the blood. RESULTS Regardless of the irradiation type, brain irradiation resulted in a notable elevation in the plasma levels of IFN-γ and MCP-1. The use of either X-ray or proton beam had differential effect on plasma cytokine levels following brain irradiation. Specifically, X-ray irradiation was associated with significantly increased plasma levels of IFN-β, IL-12p70, and IL-23, along with a decreased level of IL-1α, in comparison to proton irradiation. Correlation analysis revealed distinct cytokine regulatory patterns between X-ray and proton brain irradiation. PCA highlighted the association of MCP-1, IL-6, TNF-α, IL-17A, and IFN-γ with neutrophils, monocytes, and naïve T-cells following X-ray irradiation. TNF-α and IL-23 levels correlated with naïve CD4+-cells following proton irradiation. Tree-based models demonstrated that high TNF-α level resulted in an increase in naïve T-cells, neutrophils, and monocytes, whereas low IL-6 level was associated with decreases in these cell counts. CONCLUSION Our findings revealed distinct inflammatory responses induced by X-ray irradiation in contrast to proton brain irradiation, as demonstrated by the differential regulation of cytokines in the bloodstream. Moreover, the study highlighted the association between specific cytokine levels and various leukocyte subpopulations. Further investigation is essential to accurately determine the impact of proton and X-ray brain irradiation on the inflammatory response and the efficacy of radiotherapy treatment.
Collapse
Affiliation(s)
- Thao-Nguyen Pham
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, Caen, France; Laboratoire de physique corpusculaire, UMR6534 IN2P3/EnsiCaen, Caen, France
| | - Julie Coupey
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, Caen, France
| | - Viktoriia Ivanova
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, Caen, France
| | - Juliette Thariat
- Laboratoire de physique corpusculaire, UMR6534 IN2P3/EnsiCaen, Caen, France; Department of Radiation Oncology, Centre François-Baclesse, Caen, France.
| | - Samuel Valable
- Université de Caen Normandie, CNRS, Normandie Université, ISTCT UMR6030, GIP CYCERON, Caen, France.
| |
Collapse
|
3
|
Drayson OGG, Melemenidis S, Katila N, Viswanathan V, Kramár EA, Zhang R, Kim R, Ru N, Petit B, Dutt S, Manjappa R, Ramish Ashraf M, Lau B, Soto L, Skinner L, Yu AS, Surucu M, Maxim PG, Zebadua-Ballasteros P, Wood MA, Montay-Gruel P, Baulch JE, Vozenin MC, Loo BW, Limoli CL. A multi-institutional study to investigate the sparing effect after whole brain electron FLASH in mice: Reproducibility and temporal evolution of functional, electrophysiological, and neurogenic endpoints. Radiother Oncol 2024; 201:110534. [PMID: 39293721 DOI: 10.1016/j.radonc.2024.110534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND AND PURPOSE Ultra-high dose-rate radiotherapy (FLASH) has been shown to mitigate normal tissue toxicities associated with conventional dose rate radiotherapy (CONV) without compromising tumor killing in preclinical models. A prominent challenge in preclinical radiation research, including FLASH, is validating both the physical dosimetry and the biological effects across multiple institutions. MATERIALS AND METHODS We previously demonstrated dosimetric reproducibility of two different electron FLASH devices at separate institutions using standardized phantoms and dosimeters. In this study, tumor-free adult female mice were given 10 Gy whole brain FLASH and CONV irradiation at both institutions and evaluated for the reproducibility and temporal evolution of multiple neurobiological endpoints. RESULTS FLASH sparing of behavioral performance on novel object recognition (4 months post-irradiation) and of electrophysiologic long-term potentiation (LTP, 5 months post-irradiation) was reproduced between institutions. Differences between FLASH and CONV on the endpoints of hippocampal neurogenesis (Sox2, doublecortin), neuroinflammation (microglial activation), and electrophysiology (LTP) were not observed at early times (48 h to 2 weeks), but recovery of immature neurons by 3 weeks was greater with FLASH. CONCLUSION In summary, we demonstrated reproducible FLASH sparing effects on the brain between two different beams at two different institutions with validated dosimetry. FLASH sparing effects on the endpoints evaluated manifested at later but not the earliest time points.
Collapse
Affiliation(s)
- Olivia G G Drayson
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Stavros Melemenidis
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nikita Katila
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vignesh Viswanathan
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Enikö A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Richard Zhang
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Rachel Kim
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Benoit Petit
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Suparna Dutt
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rakesh Manjappa
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M Ramish Ashraf
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Brianna Lau
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Luis Soto
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lawrie Skinner
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amu S Yu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Murat Surucu
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter G Maxim
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Paola Zebadua-Ballasteros
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Laboratorio de Fisica Medica, Instituto Nacional de Neurología y Neurocirugía MVS, México City 14269, Mexico
| | - Marcelo A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Pierre Montay-Gruel
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland; Radiation Oncology Department, Iridium Netwerk, Wilrijk, Antwerp, Belgium; Antwerp Research in Radiation Oncology (AReRO), Centre for Oncological Research (CORE), University of Antwerp, Antwerp, Belgium
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Department of Radiation Oncology, Lausanne University Hospital and University of Lausanne, Switzerland.
| | - Billy W Loo
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
4
|
Dalboni da Rocha JL, Zou Stinnett P, Scoggins MA, McAfee SS, Conklin HM, Gajjar A, Sitaram R. Functional MRI Assessment of Brain Activity Patterns Associated with Reading in Medulloblastoma Survivors. Brain Sci 2024; 14:904. [PMID: 39335401 PMCID: PMC11429556 DOI: 10.3390/brainsci14090904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Medulloblastoma, a malignant brain tumor primarily affecting children, poses significant challenges to patients and clinicians due to its complex treatment and potential long-term cognitive consequences. While recent advancements in treatment have significantly improved survival rates, survivors often face cognitive impairments, particularly in reading, impacting their quality of life. According to the double deficit theory, reading impairments are caused by deficits in one or both of two independent reading-related functions: phonological awareness and rapid visual naming. This longitudinal study investigates neurofunctional changes related to reading in medulloblastoma survivors in comparison to controls using functional MRI acquired during rapid automatized naming tasks over three annual visits. Support vector machine classification of functional MRI data reveals a progressive divergence in brain activity patterns between medulloblastoma survivors and healthy controls over time, suggesting delayed effects of cancer treatment on brain function. Alterations in brain regions involved in visual processing and orthographic recognition during rapid naming tasks imply disruptions in the ventral visual pathway associated with normal orthographic processing. These alterations are correlated with performance in tasks involving sound awareness, reading fluency, and word attack. These findings underscore the dynamic nature of post-treatment neurofunctional alterations and the importance of early identification and intervention to address cognitive deficits in survivors.
Collapse
Affiliation(s)
- Josue L. Dalboni da Rocha
- Department of Diagnostic Imaging, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (J.L.D.d.R.); (P.Z.S.); (S.S.M.)
| | - Ping Zou Stinnett
- Department of Diagnostic Imaging, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (J.L.D.d.R.); (P.Z.S.); (S.S.M.)
| | - Matthew A. Scoggins
- Department of Psychology and Biobehavioral Sciences, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (M.A.S.); (H.M.C.)
| | - Samuel S. McAfee
- Department of Diagnostic Imaging, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (J.L.D.d.R.); (P.Z.S.); (S.S.M.)
| | - Heather M. Conklin
- Department of Psychology and Biobehavioral Sciences, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (M.A.S.); (H.M.C.)
| | - Amar Gajjar
- Department of Pediatric Medicine, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA;
| | - Ranganatha Sitaram
- Department of Diagnostic Imaging, St. Jude Children Research’s Hospital, Memphis, TN 38105, USA; (J.L.D.d.R.); (P.Z.S.); (S.S.M.)
| |
Collapse
|
5
|
Lan J, Ren Y, Liu Y, Chen L, Liu J. A bibliometric analysis of radiation-induced brain injury: a research of the literature from 1998 to 2023. Discov Oncol 2024; 15:364. [PMID: 39172266 PMCID: PMC11341524 DOI: 10.1007/s12672-024-01223-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury (RIBI) is a debilitating sequela after cranial radiotherapy. Research on the topic of RIBI has gradually entered the public eye, with more innovations and applications of evidence-based research and biological mechanism research in the field of that. This was the first bibliometric analysis on RIBI, assessing brain injury related to radiation articles that were published during 1998-2023, to provide an emerging theoretical basis for the future development of RIBI. METHODS Literature were obtained from the Web of Science Core Collection (WOSCC) from its inception to December 31, 2023. The column of publications, author details, affiliated institutions and countries, publication year, and keywords were also recorded. RESULTS A total of 2543 journal articles were selected. The annual publications on RIBI fluctuated within a certain range. Journal of Neuro-oncology was the most published journal and Radiation Oncology was the most impactful one. LIMOLI CL was the most prolific author with 37 articles and shared the highest h-index with BARNETT GH. The top one country and institutions were the USA and the University of California System, respectively. Clusters analysis of co-keywords demonstrated that the temporal research trends in this field primarily focused on imaging examination and therapy for RIBI. CONCLUSION This study collects, visualizes, and analyzes the literature within the field of RIBI over the last 25 years to map the development process, research frontiers and hotspots, and cutting-edge directions in clinical practice and mechanisms related to RIBI.
Collapse
Affiliation(s)
- Jinxin Lan
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yifan Ren
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yuyang Liu
- Department of Neurosurgery, The 920th Hospital of Joint Logistics Support Force, Kunming, 650032, Yunnan, China
| | - Ling Chen
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Jialin Liu
- Department of Neurosurgery, The First Medical Center, The Chinese PLA General Hospital, Beijing, 100853, China.
- Chinese PLA General Hospital, Chinese PLA Institute of Neurosurgery, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
6
|
Ayoub R, Yang S, Ji H, Fan L, De Michino S, Mabbott DJ, Nieman BJ. Brain volume and microglial density changes are correlated in a juvenile mouse model of cranial radiation and CSF1R inhibitor treatment. NMR IN BIOMEDICINE 2024:e5222. [PMID: 39164196 DOI: 10.1002/nbm.5222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 08/22/2024]
Abstract
Microglia have been shown to proliferate and become activated following cranial radiotherapy (CRT), resulting in a chronic inflammatory response. We investigated the role of microglia in contributing to widespread volume losses observed in the brain following CRT in juvenile mice. To manipulate microglia, we used low-dose treatment with a highly selective CSF1R inhibitor called PLX5622 (PLX). We hypothesized that alteration of the post-CRT microglia population would lead to changes in brain development outcomes, as evaluated by structural MRI. Wild-type C57BL/6J mice were provided with daily intraperitoneal injections of PLX (25 mg/kg) or vehicle from postnatal day (P)14 to P19. Mice also received whole-brain irradiation (7 Gy) or sham irradiation (0 Gy) at 16 days of age. In one cohort of mice, immunohistochemical assessment in tissue sections was conducted to assess the impact of the selected PLX and CRT doses as well as their combination. In a separate cohort, mice were imaged using MRI at P14 (pretreatment), P19, P23, P42 and P63 in order to assess induced volume changes, which were measured based on structures from a predefined atlas. We observed that PLX and radiation treatments led to sex-specific changes in the microglial cell population. Across treatment groups, MRI-detected anatomical volumes at P19 and P63 were associated with microglia and proliferating microglia densities, respectively. Overall, our study demonstrates that low-dose PLX treatment produces a sex-dependent response in juvenile mice, that manipulation of microglia alters CRT-induced volume changes and that microglia density and MRI-derived volume changes are correlated in this model.
Collapse
Affiliation(s)
- Ramy Ayoub
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sabrina Yang
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Helen Ji
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lloyd Fan
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Steven De Michino
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Donald J Mabbott
- Department of Psychology, University of Toronto, Toronto, Ontario, Canada
- Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian J Nieman
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Rodgers LT, Villano JL, Hartz AMS, Bauer B. Glioblastoma Standard of Care: Effects on Tumor Evolution and Reverse Translation in Preclinical Models. Cancers (Basel) 2024; 16:2638. [PMID: 39123366 PMCID: PMC11311277 DOI: 10.3390/cancers16152638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Glioblastoma (GBM) presents a significant public health challenge as the deadliest and most common malignant brain tumor in adults. Despite standard-of-care treatment, which includes surgery, radiation, and chemotherapy, mortality rates are high, underscoring the critical need for advancing GBM therapy. Over the past two decades, numerous clinical trials have been performed, yet only a small fraction demonstrated a benefit, raising concerns about the predictability of current preclinical models. Traditionally, preclinical studies utilize treatment-naïve tumors, failing to model the clinical scenario where patients undergo standard-of-care treatment prior to recurrence. Recurrent GBM generally exhibits distinct molecular alterations influenced by treatment selection pressures. In this review, we discuss the impact of treatment-surgery, radiation, and chemotherapy-on GBM. We also provide a summary of treatments used in preclinical models, advocating for their integration to enhance the translation of novel strategies to improve therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Louis T. Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - John L. Villano
- Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Medicine, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Neurosurgery, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Anika M. S. Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
8
|
Zhong K, Liu K, Song Y, Chen S, Hu X, Xue R, Ma X, Li S, Yang J, Deng Z, Zhu X, Yuan M, Huang Y, Yin W, Chen Y, Tang Y, Shi Z. A Synthetic Steroid 5α-Androst-3β, 5, 6β-triol Alleviates Radiation-Induced Brain Injury in Mice via Inhibiting GBP5/NF-κB/NLRP3 Signal Axis. Mol Neurobiol 2024; 61:4074-4089. [PMID: 38057643 DOI: 10.1007/s12035-023-03831-9] [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: 09/26/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy for head and neck tumors can lead to a severe complication known as radiation-induced brain injury (RIBI). However, the underlying mechanism of RIBI development remains unclear, and limited prevention and treatment options are available. Neuroactive steroids have shown potential in treating neurological disorders. 5α-Androst-3β, 5, 6β-triol (TRIOL), a synthetic neuroprotective steroid, holds promise as a treatment candidate for RIBI patients. However, the neuroprotective effects and underlying mechanism of TRIOL on RIBI treatment are yet to be elucidated. In the present study, our findings demonstrate TRIOL's potential as a neuroprotective agent against RIBI. In gamma knife irradiation mouse model, TRIOL treatment significantly reduced brain necrosis volume, microglial activation, and neuronal loss. RNA-sequencing, immunofluorescence, real-time quantitative polymerase chain reaction, siRNA transfection, and western blotting techniques revealed that TRIOL effectively decreased microglial activation, proinflammatory cytokine release, neuron loss, and guanylate-binding protein 5 (GBP5) expression, along with its downstream signaling pathways NF-κB and NLRP3 activation in vitro. In summary, TRIOL effectively alleviate RIBI by inhibiting the GBP5/NF-κB/NLRP3 signal axis, reducing microglia activation and pro-inflammation cytokines release, rescuing neuron loss. This study highlights the potential of TRIOL as a novel and promising therapy drug for RIBI treatment.
Collapse
Affiliation(s)
- Ke Zhong
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Pharmacy, Sun-Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kejia Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu Song
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Sitai Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xia Hu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ruiqi Xue
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xueying Ma
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jingwen Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaoqiu Zhu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Mingjun Yuan
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Yijun Huang
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Wei Yin
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Yupin Chen
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
9
|
Wang Y, Tian J, Liu D, Li T, Mao Y, Zhu C. Microglia in radiation-induced brain injury: Cellular and molecular mechanisms and therapeutic potential. CNS Neurosci Ther 2024; 30:e14794. [PMID: 38867379 PMCID: PMC11168970 DOI: 10.1111/cns.14794] [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: 11/23/2023] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Radiation-induced brain injury is a neurological condition resulting from radiotherapy for malignant tumors, with its underlying pathogenesis still not fully understood. Current hypotheses suggest that immune cells, particularly the excessive activation of microglia in the central nervous system and the migration of peripheral immune cells into the brain, play a critical role in initiating and progressing the injury. This review aimed to summarize the latest advances in the cellular and molecular mechanisms and the therapeutic potential of microglia in radiation-induced brain injury. METHODS This article critically examines recent developments in understanding the role of microglia activation in radiation-induced brain injury. It elucidates associated mechanisms and explores novel research pathways and therapeutic options for managing this condition. RESULTS Post-irradiation, activated microglia release numerous inflammatory factors, exacerbating neuroinflammation and facilitating the onset and progression of radiation-induced damage. Therefore, controlling microglial activation and suppressing the secretion of related inflammatory factors is crucial for preventing radiation-induced brain injury. While microglial activation is a primary factor in neuroinflammation, the precise mechanisms by which radiation prompts this activation remain elusive. Multiple signaling pathways likely contribute to microglial activation and the progression of radiation-induced brain injury. CONCLUSIONS The intricate microenvironment and molecular mechanisms associated with radiation-induced brain injury underscore the crucial roles of immune cells in its onset and progression. By investigating the interplay among microglia, neurons, astrocytes, and peripheral immune cells, potential strategies emerge to mitigate microglial activation, reduce the release of inflammatory agents, and impede the entry of peripheral immune cells into the brain.
Collapse
Affiliation(s)
- Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Jiayu Tian
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Dandan Liu
- Department of Electrocardiogram, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Tao Li
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Department of PediatricsHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Yanna Mao
- Department of Hematology and Oncology, Children's Hospital Affiliated to Zhengzhou UniversityHenan Children's Hospital Zhengzhou Children's HospitalZhengzhouChina
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Department of PediatricsInstitute of Neuroscience and Third Affiliated Hospital of Zhengzhou UniversityKangfuqian Street 7Zhengzhou450052None SelectedChina
- Center for Brain Repair and Rehabilitation, Department of Clinical NeuroscienceInstitute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgMedicinaregtan 11Göteborg40530Sweden
| |
Collapse
|
10
|
Demos-Davies K, Lawrence J, Coffey J, Morgan A, Ferreira C, Hoeppner LH, Seelig D. Longitudinal Neuropathological Consequences of Extracranial Radiation Therapy in Mice. Int J Mol Sci 2024; 25:5731. [PMID: 38891920 PMCID: PMC11171684 DOI: 10.3390/ijms25115731] [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: 04/18/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer-related cognitive impairment (CRCI) is a consequence of chemotherapy and extracranial radiation therapy (ECRT). Our prior work demonstrated gliosis in the brain following ECRT in SKH1 mice. The signals that induce gliosis were unclear. Right hindlimb skin from SKH1 mice was treated with 20 Gy or 30 Gy to induce subclinical or clinical dermatitis, respectively. Mice were euthanized at 6 h, 24 h, 5 days, 12 days, and 25 days post irradiation, and the brain, thoracic spinal cord, and skin were collected. The brains were harvested for spatial proteomics, immunohistochemistry, Nanostring nCounter® glial profiling, and neuroinflammation gene panels. The thoracic spinal cords were evaluated by immunohistochemistry. Radiation injury to the skin was evaluated by histology. The genes associated with neurotransmission, glial cell activation, innate immune signaling, cell signal transduction, and cancer were differentially expressed in the brains from mice treated with ECRT compared to the controls. Dose-dependent increases in neuroinflammatory-associated and neurodegenerative-disease-associated proteins were measured in the brains from ECRT-treated mice. Histologic changes in the ECRT-treated mice included acute dermatitis within the irradiated skin of the hindlimb and astrocyte activation within the thoracic spinal cord. Collectively, these findings highlight indirect neuronal transmission and glial cell activation in the pathogenesis of ECRT-related CRCI, providing possible signaling pathways for mitigation strategies.
Collapse
Affiliation(s)
- Kimberly Demos-Davies
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Jessica Coffey
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Amy Morgan
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
| | - Clara Ferreira
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Luke H. Hoeppner
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Davis Seelig
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (J.C.); (A.M.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
11
|
Ma T, Li K, Sang W, Liu X, Luo Q, Peng Y, Wang M, Luo X, Fang J, Wang H, Wang T, Zuo C. Low-dose-rate induces more severe cognitive impairment than high-dose-rate in rats exposed to chronic low-dose γ-radiation. Front Public Health 2024; 12:1387330. [PMID: 38841686 PMCID: PMC11150688 DOI: 10.3389/fpubh.2024.1387330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
Background Owing to the long penetration depth of gamma (γ)-rays, individuals working in ionizing radiation environments are chronically exposed to low-dose γ-radiation, resulting in cognitive changes. Dose rate significantly affects radiation-induced biological effects; however, its role in chronic low-dose γ-irradiation-induced cognitive impairment remains unclear. We aimed to investigate whether chronic low-dose γ-irradiation at low-dose-rate (LDR) could induce cognitive impairment and to compare the cognitive alteration caused by chronic low-dose γ-irradiation at LDR and high-dose-rate (HDR). Methods The rats were exposed to γ-irradiation at a LDR of 6 mGy/h and a HDR of 20 mGy/h for 30 days (5 h/day). Functional imaging was performed to assess the brain inflammation and blood-brain barrier (BBB) destruction of rats. Histological and immunofluorescence analyses were used to reveal the neuron damage and the activation of microglia and astrocytes in the hippocampus. RNA sequencing was conducted to investigate changes in gene expression in hippocampus. Results The rats in the LDR group exhibited more persistent cognitive impairment than those in the HDR group. Furthermore, irradiated rats showed brain inflammation and a compromised BBB. Histologically, the number of hippocampal neurons were comparable in the LDR group but were markedly decreased in the HDR. Additionally, activated M1-like microglia and A1-like astrocytes were observed in the hippocampus of rats in the LDR group; however, only M1-like microglia were activated in the HDR group. Mechanistically, the PI3K-Akt signaling pathway contributed to the different cognitive function change between the LDR group and HDR group. Conclusion Compared with chronic low-dose γ-irradiation at HDR, LDR induced more severe cognitive impairment which might involve PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Tianbao Ma
- School of Medicine, Shanghai University, Shanghai, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Kexian Li
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Wenjuan Sang
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Xingyu Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Qun Luo
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Ye Peng
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Mingxing Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiu Luo
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jingjing Fang
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Haijun Wang
- Naval Medical Center, Naval Medical University, Shanghai, China
| | - Tao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Changjing Zuo
- School of Medicine, Shanghai University, Shanghai, China
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
12
|
Vāvere AL, Ghosh A, Amador Diaz V, Clay AJ, Hall PM, Neumann KD. Automated radiosynthesis of [ 18F]DPA-714 on a commercially available IBA Synthera®. Appl Radiat Isot 2024; 207:111257. [PMID: 38461627 PMCID: PMC10984111 DOI: 10.1016/j.apradiso.2024.111257] [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: 11/28/2023] [Revised: 02/08/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024]
Abstract
The goal of this work was to develop a reliable method to produce the well-validated microglial activation PET tracer, [18F]DPA-714, routinely for clinical and preclinical research using an IBA Synthera®. Optimization of literature methods included reduced precursor mass and use of TBA HCO3 as the phase transfer agent in place of Kryptofix® 222 in a 65-min synthesis with an average activity yield of 24.6 ± 3.8% (n = 5). Successful quality control testing and process validation results are reported.
Collapse
Affiliation(s)
- Amy L Vāvere
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Arijit Ghosh
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Victor Amador Diaz
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Allison J Clay
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter M Hall
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kiel D Neumann
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
13
|
Praveen Kumar C, Aggarwal LM, Bhasi S, Sharma N. A Monte Carlo simulation-based decision support system for radiation oncologists in the treatment of glioblastoma multiforme. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2024; 63:215-262. [PMID: 38664268 DOI: 10.1007/s00411-024-01065-4] [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: 09/07/2021] [Accepted: 03/24/2024] [Indexed: 05/15/2024]
Abstract
In the present research, we have developed a model-based crisp logic function statistical classifier decision support system supplemented with treatment planning systems for radiation oncologists in the treatment of glioblastoma multiforme (GBM). This system is based on Monte Carlo radiation transport simulation and it recreates visualization of treatment environments on mathematical anthropomorphic brain (MAB) phantoms. Energy deposition within tumour tissue and normal tissues are graded by quality audit factors which ensure planned dose delivery to tumour site thereby minimising damages to healthy tissues. The proposed novel methodology predicts tumour growth response to radiation therapy from a patient-specific medicine quality audit perspective. Validation of the study was achieved by recreating thirty-eight patient-specific mathematical anthropomorphic brain phantoms of treatment environments by taking into consideration density variation and composition of brain tissues. Dose computations accomplished through water phantom, tissue-equivalent head phantoms are neither cost-effective, nor patient-specific customized and is often less accurate. The above-highlighted drawbacks can be overcome by using open-source Electron Gamma Shower (EGSnrc) software and clinical case reports for MAB phantom synthesis which would result in accurate dosimetry with due consideration to the time factors. Considerable dose deviations occur at the tumour site for environments with intraventricular glioblastoma, haematoma, abscess, trapped air and cranial flaps leading to quality factors with a lower logic value of 0. Logic value of 1 depicts higher dose deposition within healthy tissues and also leptomeninges for majority of the environments which results in radiation-induced laceration.
Collapse
Affiliation(s)
- C Praveen Kumar
- School of Biomedical Engineering, Indian Institute of Technology - BHU, Varanasi, India.
| | - Lalit M Aggarwal
- Department of Radiotherapy, Institute of Medical Sciences - BHU, Varanasi, India
| | - Saju Bhasi
- Division of Radiation Physics, Regional Cancer Centre, Thiruvananthapuram, India
| | - Neeraj Sharma
- School of Biomedical Engineering, Indian Institute of Technology - BHU, Varanasi, India
| |
Collapse
|
14
|
Elmas O, Keskin E, Keser Sahin HH, Guven B, Almisned G, Zakaly HM, Tekin H, Ene A. The effect of Annona muricata (Graviola) on the prevention of brain damage due to ionizing radiation in rats. Heliyon 2024; 10:e25932. [PMID: 38370260 PMCID: PMC10867642 DOI: 10.1016/j.heliyon.2024.e25932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 01/03/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024] Open
Abstract
In this study, it was aimed to evaluate the effect of ethanol extract of Annona Muricata (AM) leaves in the prevention of brain damage caused by ionizing radiation (IR). This study was conducted in the Experimental Animal Research Unit of a university with 28 adults female Wistar Albino rats. The experimental groups were as follows: Control group (n = 8), AM group (n = 6), IR group (n = 8), AM + IR group (n = 6). In the IR group, astrocyte hypertrophy, microglial reaction and inflammatory reaction levels were significantly higher than the control and AM groups (P < 0.001). Edema was significantly higher in the IR group compared to the control group (P=0.001). The MDA of the IR group was significantly higher compared to the control group and AM group (P=0.031, P=0.006, respectively). The MDA of the AM + IR group was significantly higher than the AM group (P=0.039). Our findings show that histomorphology and oxidant damage caused by IR can be ameliorated using AM, as demonstrated by the comparison of the controls to AM + IR recipients, which showed similar histomorphology and oxidant damage levels.
Collapse
Affiliation(s)
- Ozlem Elmas
- Department of Radiation Oncology, Bulent Ecevit University Practice and Research Hospital, Zonguldak, Turkey
| | - Emrah Keskin
- Department of Neurosurgery, Bulent Ecevit University Practice and Research Hospital, Zonguldak, Turkey
| | | | - Berrak Guven
- Department of Biochemistry, Bulent Ecevit University Practice and Research Hospital, Zonguldak, Turkey
| | - Ghada Almisned
- Department of Physics, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hesham M.H. Zakaly
- Institute of Physics and Technology, Ural Federal University, Yekaterinburg, 620002, Russia
- Physics Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - H.O. Tekin
- Department of Medical Diagnostic Imaging, College of Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates
- Istinye University, Faculty of Engineering and Natural Sciences, Computer Engineering Department, Istanbul, 34396, Turkey
| | - Antoaneta Ene
- INPOLDE Research Center, Department of Chemistry, Physics and Environment, Faculty of Sciences and Environment, Dunarea de Jos University of Galati, 47 Domneasca Street, 800008 Galati, Romania
| |
Collapse
|
15
|
Voshart DC, Oshima T, Jiang Y, van der Linden GP, Ainslie AP, Reali Nazario L, van Buuren-Broek F, Scholma AC, van Weering HRJ, Brouwer N, Sewdihal J, Brouwer U, Coppes RP, Holtman IR, Eggen BJL, Kooistra SM, Barazzuol L. Radiotherapy induces persistent innate immune reprogramming of microglia into a primed state. Cell Rep 2024; 43:113764. [PMID: 38358885 DOI: 10.1016/j.celrep.2024.113764] [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: 05/16/2023] [Revised: 12/06/2023] [Accepted: 01/25/2024] [Indexed: 02/17/2024] Open
Abstract
Over half of patients with brain tumors experience debilitating and often progressive cognitive decline after radiotherapy treatment. Microglia, the resident macrophages in the brain, have been implicated in this decline. In response to various insults, microglia can develop innate immune memory (IIM), which can either enhance (priming or training) or repress (tolerance) the response to subsequent inflammatory challenges. Here, we investigate whether radiation affects the IIM of microglia by irradiating the brains of rats and later exposing them to a secondary inflammatory stimulus. Comparative transcriptomic profiling and protein validation of microglia isolated from irradiated rats show a stronger immune response to a secondary inflammatory insult, demonstrating that radiation can lead to long-lasting molecular reprogramming of microglia. Transcriptomic analysis of postmortem normal-appearing non-tumor brain tissue of patients with glioblastoma indicates that radiation-induced microglial priming is likely conserved in humans. Targeting microglial priming or avoiding further inflammatory insults could decrease radiotherapy-induced neurotoxicity.
Collapse
Affiliation(s)
- Daniëlle C Voshart
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Takuya Oshima
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Yuting Jiang
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Gideon P van der Linden
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Anna P Ainslie
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands; European Research Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Luiza Reali Nazario
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Fleur van Buuren-Broek
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Ayla C Scholma
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Hilmar R J van Weering
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Jeffrey Sewdihal
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Uilke Brouwer
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Rob P Coppes
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences, Section of Molecular Neurobiology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands
| | - Lara Barazzuol
- Department of Biomedical Sciences, Section of Molecular Cell Biology, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, the Netherlands; Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands.
| |
Collapse
|
16
|
Wu B, Li S, Wang J, Wang J, Qiu W, Gao H. Bibliometric and visualization analysis of radiation brain injury from 2003 to 2023. Front Neurol 2024; 14:1275836. [PMID: 38298563 PMCID: PMC10828967 DOI: 10.3389/fneur.2023.1275836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/15/2023] [Indexed: 02/02/2024] Open
Abstract
Background Over the past two decades, the field of radiation brain injury has attracted the attention of an increasing number of brain scientists, particularly in the areas of molecular pathology and therapeutic approaches. Characterizing global collaboration networks and mapping development trends over the past 20 years is essential. Objective The aim of this paper is to examine significant issues and future directions while shedding light on collaboration and research status in the field of radiation brain injury. Methods Bibliometric studies were performed using CiteSpaceR-bibliometrix and VOSviewer software on papers regarding radiation brain injury that were published before November 2023 in the Web of Science Core Collection. Results In the final analysis, we found 4,913 records written in 1,219 publications by 21,529 authors from 5,007 institutions in 75 countries. There was a noticeable increase in publications in 2014 and 2021. The majority of records listed were produced by China, the United States, and other high-income countries. The largest nodes in each cluster of the collaboration network were Sun Yat-sen University, University of California-San Francisco, and the University of Toronto. Galldiks N, Barnett GH, Langen KJ and Kim JH are known to be core authors in the field. The top 3 keywords in that time frame are radiation, radiation necrosis, and radiation-therapy. Conclusions The objective and thorough bibliometric analysis also identifies current research hotspots and potential future paths, providing a retrospective perspective on RBI and offering useful advice to researchers choosing research topics. Future development directions include the integration of multi-omics methodologies and novel imaging techniques to improve RBI's diagnostic effectiveness and the search for new therapeutic targets.
Collapse
Affiliation(s)
- Baofang Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Shaojie Li
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Jian Wang
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiayin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Weizhi Qiu
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Department of Neurosurgery, The Second Affiliated Clinical Medical College of Fujian Medical University, Quanzhou, China
| |
Collapse
|
17
|
Schumacher S, Tahiri H, Ezan P, Rouach N, Witschas K, Leybaert L. Inhibiting astrocyte connexin-43 hemichannels blocks radiation-induced vesicular VEGF-A release and blood-brain barrier dysfunction. Glia 2024; 72:34-50. [PMID: 37670489 DOI: 10.1002/glia.24460] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 09/07/2023]
Abstract
Therapeutic brain irradiation with ionizing radiation exerts multiple side effects including barrier leakage that disturbs glial-neuronal functioning and may affect cognition. Astrocytes contribute to barrier leakage by endfeet release of various vasoactive substances acting on capillary endothelial cells forming the barrier. Here, we investigated X-ray effects on astrocytic vesicular transport in mice and determined whether interfering with astrocyte connexins affects radiation-induced barrier leakage. We found that astrocytic VEGF-A-loaded VAMP3 vesicles drastically reorganize starting from 6 h post-irradiation and move in a calcium- and Cx43-dependent manner towards endfeet where VEGF-A is released, provoking barrier leakage. Vesicular transport activation, VEGF-A release and leakage 24 h post-irradiation were all potently inhibited by astrocytic Cx43 KO, Cx43S255/262/279/282A (MK4) mutant mice and TATGap19 inhibition of Cx43 hemichannel opening. Astrocyte VEGF release is a major player in complications of brain irradiation, which can be mitigated by anti-VEGF treatments. Targeting Cx43 hemichannels allows to prevent astrocyte VEGF release at an early stage after brain irradiation.
Collapse
Affiliation(s)
- Steffi Schumacher
- Department of Basic and Applied Medical Sciences - Physiology Group, Ghent University, Ghent, Belgium
| | - Hanane Tahiri
- Department of Basic and Applied Medical Sciences - Physiology Group, Ghent University, Ghent, Belgium
| | - Pascal Ezan
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Labex Memolife, Paris, France
| | - Katja Witschas
- Department of Basic and Applied Medical Sciences - Physiology Group, Ghent University, Ghent, Belgium
| | - Luc Leybaert
- Department of Basic and Applied Medical Sciences - Physiology Group, Ghent University, Ghent, Belgium
| |
Collapse
|
18
|
Demos-Davies K, Lawrence J, Ferreira C, Seelig D. The Distant Molecular Effects on the Brain by Cancer Treatment. Brain Sci 2023; 14:22. [PMID: 38248237 PMCID: PMC10813787 DOI: 10.3390/brainsci14010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer survivors experience cancer-related cognitive impairment (CRCI) secondary to treatment. Chemotherapy and radiation therapy independently contribute to cognitive dysfunction; however, the underlying mechanisms leading to dysfunction remain unclear. We characterized brain gene expression changes in a mouse model of CRCI to identify the mechanistic underpinnings. Eleven-to-twelve-week-old SKH1 mice were treated with doxorubicin (DOX), hindlimb radiation (RT), concurrent hindlimb radiation and doxorubicin (DOX-RT), or no treatment (control). Sixteen days following treatment, gene expression was measured from murine brains using the NanoString nCounter® glial profiling panel. Gene expression was normalized and compared between groups. No two groups shared the same expression pattern, and only Gnb1 and Srpr were upregulated in multiple treatment groups. Brains from DOX-treated mice had upregulated Atf2, Atp5b, Gnb1, Rad23b, and Srpr and downregulated Sirt5 expression compared to control brains. Brains from RT-treated mice demonstrated increased Abcg2 and Fgf2 and decreased C1qa and C1qb expression compared to control brains. Brains from DOX-RT-treated mice had upregulated Adar, E2f3, Erlec1, Gnb1, Srpr, Vim, and Pdgfra expression and downregulated Rock2 and Inpp5f expression compared to control brains. The gene expression changes demonstrated here highlight roles for neuronal transmission and oxidative stress in the pathogenesis of doxorubicin-related CRCI and inflammation in RT-related CRCI.
Collapse
Affiliation(s)
- Kimberly Demos-Davies
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (D.S.)
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455,USA
| | - Clara Ferreira
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Davis Seelig
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, Saint Paul, MN 55108, USA; (J.L.); (D.S.)
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455,USA
| |
Collapse
|
19
|
Ye Z, Wang J, Shi W, Zhou Z, Zhang Y, Wang J, Yang H. Reprimo (RPRM) as a Potential Preventive and Therapeutic Target for Radiation-Induced Brain Injury via Multiple Mechanisms. Int J Mol Sci 2023; 24:17055. [PMID: 38069378 PMCID: PMC10707327 DOI: 10.3390/ijms242317055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Patients receiving cranial radiotherapy for primary and metastatic brain tumors may experience radiation-induced brain injury (RIBI). Thus far, there has been a lack of effective preventive and therapeutic strategies for RIBI. Due to its complicated underlying pathogenic mechanisms, it is rather difficult to develop a single approach to target them simultaneously. We have recently reported that Reprimo (RPRM), a tumor suppressor gene, is a critical player in DNA damage repair, and RPRM deletion significantly confers radioresistance to mice. Herein, by using an RPRM knockout (KO) mouse model established in our laboratory, we found that RPRM deletion alleviated RIBI in mice via targeting its multiple underlying mechanisms. Specifically, RPRM knockout significantly reduced hippocampal DNA damage and apoptosis shortly after mice were exposed to whole-brain irradiation (WBI). For the late-delayed effect of WBI, RPRM knockout obviously ameliorated a radiation-induced decline in neurocognitive function and dramatically diminished WBI-induced neurogenesis inhibition. Moreover, RPRM KO mice exhibited a significantly lower level of acute and chronic inflammation response and microglial activation than wild-type (WT) mice post-WBI. Finally, we uncovered that RPRM knockout not only protected microglia against radiation-induced damage, thus preventing microglial activation, but also protected neurons and decreased the induction of CCL2 in neurons after irradiation, in turn attenuating the activation of microglial cells nearby through paracrine CCL2. Taken together, our results indicate that RPRM plays a crucial role in the occurrence of RIBI, suggesting that RPRM may serve as a novel potential target for the prevention and treatment of RIBI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongying Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Suzhou Medical College of Soochow University, Suzhou 215123, China; (Z.Y.); (J.W.); (W.S.); (Z.Z.); (Y.Z.); (J.W.)
| |
Collapse
|
20
|
Krauss P, Kahl KH, Bonk MN, Wolfert C, Sommer B, Stueben G, Shiban E. Intraoperative radiotherapy after resection of brain metastases located in the posterior fossa. Analysis of postoperative morbidity and mortality in a single center cohort. J Clin Neurosci 2023; 118:1-6. [PMID: 37832264 DOI: 10.1016/j.jocn.2023.09.014] [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: 08/08/2023] [Revised: 09/14/2023] [Accepted: 09/16/2023] [Indexed: 10/15/2023]
Abstract
INTRODUCTION In order to improve surgical outcome and accelerate the adjuvant oncologic therapy, intraoperative Radiotherapy (IORT) has become a treatment option in oncologic surgery for various diseases including glioma and brain metastasis (BM). BMs are often located in the cranial posterior fossa (PF) requiring specific surgical considerations due to its complex anatomy. Up until now, data on IORT for BMs is limited and detailed description in the use of IORT for lesions in the PF is lacking. Our aim is to provide more insight into this emerging treatment strategy. METHODS We performed a retrospective analysis of patients receiving surgery for BMs and undergoing IORT at our institution. Each patient was discussed at the interdisciplinary tumor board decision before the intervention. Patient characteristics, functional status (Karnofsky Performance Score, KPS) before and after surgery, disease (recursive partitioning analysis, lesion size) and operative parameters were analyzed. Adverse events (AE) were recorded up until 30 days after the intervention and rated according to the Clavien Dindo Rating scale. RESULTS Nine patients (5 female) were included. None underwent prior radiotherapy (RT). Mean age was 66 ± 11 years. Preoperative median KPS was 80%. Mean BM diameter was 3.2 ± 0.9 cm. There was no statistically significant deterioration of the functional status after the intervention. Two patients experienced AEs with both of them needing revision surgery. CONCLUSION Surgery for BMs with IORT in the PF seems safe and feasible. Further studies are needed to evaluate the influence of IORT on long-term outcome after BM surgery.
Collapse
Affiliation(s)
- Philipp Krauss
- Department of Neurosurgery, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany.
| | - Klaus Henning Kahl
- Department of Radiooncology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Maximilian Niklas Bonk
- Department of Neurosurgery, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Christina Wolfert
- Department of Neurosurgery, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Bjoern Sommer
- Department of Neurosurgery, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Georg Stueben
- Department of Radiooncology, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| | - Ehab Shiban
- Department of Neurosurgery, University Hospital Augsburg, Stenglinstrasse 2, 86156 Augsburg, Germany
| |
Collapse
|
21
|
Riviere-Cazaux C, Carlstrom LP, Neth BJ, Olson IE, Rajani K, Rahman M, Ikram S, Mansour MA, Mukherjee B, Warrington AE, Short SC, von Zglinicki T, Brown DA, Burma S, Tchkonia T, Schafer MJ, Baker DJ, Kizilbash SH, Kirkland JL, Burns TC. An untapped window of opportunity for glioma: targeting therapy-induced senescence prior to recurrence. NPJ Precis Oncol 2023; 7:126. [PMID: 38030881 PMCID: PMC10687268 DOI: 10.1038/s41698-023-00476-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
High-grade gliomas are primary brain tumors that are incredibly refractory long-term to surgery and chemoradiation, with no proven durable salvage therapies for patients that have failed conventional treatments. Post-treatment, the latent glioma and its microenvironment are characterized by a senescent-like state of mitotic arrest and a senescence-associated secretory phenotype (SASP) induced by prior chemoradiation. Although senescence was once thought to be irreversible, recent evidence has demonstrated that cells may escape this state and re-enter the cell cycle, contributing to tumor recurrence. Moreover, senescent tumor cells could spur the growth of their non-senescent counterparts, thereby accelerating recurrence. In this review, we highlight emerging evidence supporting the use of senolytic agents to ablate latent, senescent-like cells that could contribute to tumor recurrence. We also discuss how senescent cell clearance can decrease the SASP within the tumor microenvironment thereby reducing tumor aggressiveness at recurrence. Finally, senolytics could improve the long-term sequelae of prior therapy on cognition and bone marrow function. We critically review the senolytic drugs currently under preclinical and clinical investigation and the potential challenges that may be associated with deploying senolytics against latent glioma. In conclusion, senescence in glioma and the microenvironment are critical and potential targets for delaying or preventing tumor recurrence and improving patient functional outcomes through senotherapeutics.
Collapse
Affiliation(s)
| | | | | | - Ian E Olson
- Department of Neurological Surgery, Northwestern University, Chicago, IL, USA
| | | | - Masum Rahman
- Department of Neurological Surgery, Rochester, MN, USA
| | - Samar Ikram
- Department of Neurological Surgery, Rochester, MN, USA
| | | | - Bipasha Mukherjee
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Arthur E Warrington
- Department of Neurological Surgery, Rochester, MN, USA
- Department of Neurology, Rochester, MN, USA
| | - Susan C Short
- Leeds Institute of Medical Research at St. James's, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Thomas von Zglinicki
- Biosciences Institute, Faculty of Medical Sciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Desmond A Brown
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Burma
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | - Marissa J Schafer
- Department of Physiology and Biomedical Engineering, Rochester, MN, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Rochester, MN, USA
| | | | - James L Kirkland
- Department of Pediatric and Adolescent Medicine, Rochester, MN, USA
- Department of Medicine, Rochester, MN, USA
| | - Terry C Burns
- Department of Neurological Surgery, Rochester, MN, USA.
| |
Collapse
|
22
|
Hardy SJ, Finkelstein A, Milano MT, Schifitto G, Sun H, Holley K, Usuki K, Weber MT, Zheng D, Seplaki CL, Janelsins M. Association of Radiation Dose to the Amygdala-Orbitofrontal Network with Emotion Recognition Task Performance in Patients with Low-Grade and Benign Brain Tumors. Cancers (Basel) 2023; 15:5544. [PMID: 38067248 PMCID: PMC10705220 DOI: 10.3390/cancers15235544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/16/2023] [Accepted: 11/18/2023] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Although data are limited, difficulty in social cognition occurs in up to 83% of patients with brain tumors. It is unknown whether cranial radiation therapy (RT) dose to the amygdala-orbitofrontal network can impact social cognition. METHODS We prospectively enrolled 51 patients with low-grade and benign brain tumors planned for cranial RT. We assessed longitudinal changes on an emotion recognition task (ERT) that measures the ability to recognize emotional states by displaying faces expressing six basic emotions and their association with the RT dose to the amygdala-orbitofrontal network. ERT outcomes included the median time to choose a response (ERTOMDRT) or correct response (ERTOMDCRT) and total correct responses (ERTHH). RESULTS The RT dose to the amygdala-orbitofrontal network was significantly associated with longer median response times on the ERT. Increases in median response times occurred at lower doses than decreases in total correct responses. The medial orbitofrontal cortex was the most important variable on regression trees predicting change in the ERTOMDCRT. DISCUSSION This is, to our knowledge, the first study to show that off-target RT dose to the amygdala-orbitofrontal network is associated with performance on a social cognition task, a facet of cognition that has previously not been mechanistically studied after cranial RT.
Collapse
Affiliation(s)
- Sara J. Hardy
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Alan Finkelstein
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14627, USA;
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael T. Milano
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Hongying Sun
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
| | - Koren Holley
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Kenneth Usuki
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Miriam T. Weber
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Dandan Zheng
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
| | - Christopher L. Seplaki
- Department of Public Health Sciences, University of Rochester Medical Center, Rochester, NY 14642, USA;
- Office for Aging Research and Health Services, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michelle Janelsins
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14620, USA; (M.T.M.); (D.Z.); (M.J.)
- Department of Surgery, Supportive Care in Cancer, University of Rochester Medical Center, Rochester, NY 14642, USA; (H.S.); (M.T.W.)
| |
Collapse
|
23
|
Lo Cascio C, Margaryan T, Luna-Melendez E, McNamara JB, White CI, Knight W, Ganta S, Opachich Z, Cantoni C, Yoo W, Sanai N, Tovmasyan A, Mehta S. Quisinostat is a brain-penetrant radiosensitizer in glioblastoma. JCI Insight 2023; 8:e167081. [PMID: 37991020 PMCID: PMC10721329 DOI: 10.1172/jci.insight.167081] [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: 11/09/2022] [Accepted: 10/13/2023] [Indexed: 11/23/2023] Open
Abstract
Histone deacetylase (HDAC) inhibitors have garnered considerable interest for the treatment of adult and pediatric malignant brain tumors. However, owing to their broad-spectrum nature and inability to effectively penetrate the blood-brain barrier, HDAC inhibitors have failed to provide substantial clinical benefit to patients with glioblastoma (GBM) to date. Moreover, global inhibition of HDACs results in widespread toxicity, highlighting the need for selective isoform targeting. Although no isoform-specific HDAC inhibitors are currently available, the second-generation hydroxamic acid-based HDAC inhibitor quisinostat possesses subnanomolar specificity for class I HDAC isoforms, particularly HDAC1 and HDAC2. It has been shown that HDAC1 is the essential HDAC in GBM. This study analyzed the neuropharmacokinetic, pharmacodynamic, and radiation-sensitizing properties of quisinostat in preclinical models of GBM. It was found that quisinostat is a well-tolerated and brain-penetrant molecule that extended survival when administered in combination with radiation in vivo. The pharmacokinetic-pharmacodynamic-efficacy relationship was established by correlating free drug concentrations and evidence of target modulation in the brain with survival benefit. Together, these data provide a strong rationale for clinical development of quisinostat as a radiosensitizer for the treatment of GBM.
Collapse
Affiliation(s)
- Costanza Lo Cascio
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Tigran Margaryan
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Ernesto Luna-Melendez
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - James B. McNamara
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Connor I. White
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - William Knight
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Saisrinidhi Ganta
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Zorana Opachich
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Claudia Cantoni
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Wonsuk Yoo
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Nader Sanai
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Artak Tovmasyan
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| | - Shwetal Mehta
- Ivy Brain Tumor Center and
- Department of Translational Neuroscience, Barrow Neurological Institute, St. Joseph’s Hospital and Medical Center, Phoenix, Arizona, USA
| |
Collapse
|
24
|
Janssen P, De Pauw L, Mambretti M, Lara O, Walckiers J, Mackens L, Rooman I, Guillaume B, De Ridder M, Ates G, Massie A. Characterization of the long-term effects of lethal total body irradiation followed by bone marrow transplantation on the brain of C57BL/6 mice. Int J Radiat Biol 2023; 100:385-398. [PMID: 37976378 DOI: 10.1080/09553002.2023.2283092] [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: 07/07/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Total body irradiation (TBI) followed by bone marrow transplantation (BMT) is used in pre-clinical research to generate mouse chimeras that allow to study the function of a protein specifically on immune cells. Adverse consequences of irradiation on the juvenile body and brain are well described and include general fatigue, neuroinflammation, neurodegeneration and cognitive impairment. Yet, the long-term consequences of TBI/BMT performed on healthy adult mice have been poorly investigated. MATERIAL AND METHODS We developed a robust protocol to achieve near complete bone marrow replacement in mice using 2x550cGy TBI and evaluated the impact of the procedure on their general health, mood disturbances, memory, brain atrophy, neurogenesis, neuroinflammation and blood-brain barrier (BBB) permeability 2 and/or 16 months post-BMT. RESULTS We found a persistent decrease in weight along with long-term impact on locomotion after TBI and BMT. Although the TBI/BMT procedure did not lead to anxiety- or depressive-like behavior 2- or 16-months post-BMT, long-term spatial memory of the irradiated mice was impaired. We also observed radiation-induced impaired neurogenesis and cortical microglia activation 2 months post-BMT. Moreover, higher levels of hippocampal IgG in aged BMT mice suggest an enhanced age-related increase in BBB permeability that could potentially contribute to the observed memory deficit. CONCLUSIONS Overall health of the mice did not seem to be majorly impacted by TBI followed by BMT during adulthood. Yet, TBI-induced alterations in the brain and behavior could lead to erroneous conclusions on the function of a protein on immune cells when comparing mouse chimeras with different genetic backgrounds that might display altered susceptibility to radiation-induced damage. Ultimately, the BMT model we here present could also be used to study the related long-term consequences of TBI and BMT seen in patients.
Collapse
Affiliation(s)
- P Janssen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Medical and Molecular Oncology, Oncology Research Centre (ORC), VUB, Brussels, Belgium
| | - L De Pauw
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - M Mambretti
- Laboratory of Medical and Molecular Oncology, Oncology Research Centre (ORC), VUB, Brussels, Belgium
| | - O Lara
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Medical and Molecular Oncology, Oncology Research Centre (ORC), VUB, Brussels, Belgium
| | - J Walckiers
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - L Mackens
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - I Rooman
- Laboratory of Medical and Molecular Oncology, Oncology Research Centre (ORC), VUB, Brussels, Belgium
| | - B Guillaume
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, UCLouvain, Brussels, Belgium
- Centre hospitalier de Jolimont, Service de Biochimie Médicale, La Louvière, Belgium
| | - M De Ridder
- Department of Radiotherapy, UZ Brussel, VUB, Brussels, Belgium
| | - G Ates
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - A Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
25
|
Hurley K, Clow R, Jadhav A, Azzam EI, Wang Y. Mitigation of acute radiation syndrome (ARS) with human umbilical cord blood. Int J Radiat Biol 2023; 100:317-334. [PMID: 37967239 DOI: 10.1080/09553002.2023.2277372] [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: 05/30/2023] [Accepted: 09/27/2023] [Indexed: 11/17/2023]
Abstract
PURPOSE The growing concern over potential unintended nuclear accidents or malicious activities involving nuclear/radiological devices cannot be overstated. Exposure to whole-body doses of radiation can result in acute radiation syndrome (ARS), colloquially known as "radiation sickness," which can severely damage various organ systems. Long-term health consequences, such as cancer and cardiovascular disease, can develop many years post-exposure. Identifying effective medical countermeasures and devising a strategic medical plan represents an urgent, unmet need. Various clinical studies have investigated the therapeutic use of umbilical cord blood (UCB) for a range of illnesses, including ARS. The objective of this review is to thoroughly discuss ARS and its sub-syndromes, and to highlight recent findings regarding the use of UCB for radiation injury. UCB, a rich source of stem cells, boasts numerous advantages over other stem cell sources, like bone marrow, owing to its ease of collection and relatively low risk of severe graft-versus-host disease. Preclinical studies suggest that treatment with UCB, and often UCB-derived mesenchymal stromal cells (MSCs), results in improved survival, accelerated hematopoietic recovery, reduced gastrointestinal tract damage, and mitigation of radiation-induced pneumonitis and pulmonary fibrosis. Interestingly, recent evidence suggests that UCB-derived exosomes and their microRNAs (miRNAs) might assist in treating radiation-induced damage, largely by inhibiting fibrotic pathways. CONCLUSION UCB holds substantial potential as a radiation countermeasure, and future research should focus on establishing treatment parameters for ARS victims.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Rachel Clow
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Ashok Jadhav
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Edouard I Azzam
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| |
Collapse
|
26
|
Sorokina SS, Malkov AE, Rozanova OM, Smirnova EN, Shemyakov AE. Behavioral performance and microglial status in mice after moderate dose of proton irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2023; 62:497-509. [PMID: 37794305 DOI: 10.1007/s00411-023-01044-1] [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: 12/30/2022] [Accepted: 09/19/2023] [Indexed: 10/06/2023]
Abstract
Cognitive impairment is a remote effect of gamma radiation treatment of malignancies. The major part of the studies on the effect of proton irradiation (a promising alternative in the treatment of radio-resistant tumors and tumors located close to critical organs) on the cognitive abilities of laboratory animals and their relation to morphological changes in the brain is rather contradictory. The aim of this study was to investigate cognitive functions and the dynamics of changes in morphological parameters of hippocampal microglial cells after 7.5 Gy of proton irradiation. Two months after the cranial irradiation, 8- to 9-week-old male SHK mice were tested for total activity, spatial learning, as well as long- and short-term hippocampus-dependent memory. To estimate the morphological parameters of microglia, brain slices of control and irradiated animals each with different time after proton irradiation (24 h, 7 days, 1 month) were stained for microglial marker Iba-1. No changes in behavior or deficits in short-term and long-term hippocampus-dependent memory were found, but an impairment of episodic memory was observed. A change in the morphology of hippocampal microglial cells, which is characteristic of the transition of cells to an activated state, was detected. One day after proton exposure in the brain tissue, a slight decrease in cell density was observed, which was restored to the control level by the 30th day after treatment. The results obtained may be promising with regard to the future use of using high doses of protons per fraction in the irradiation of tumors.
Collapse
Affiliation(s)
- S S Sorokina
- Laboratory of Isotope Investigations, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia.
| | - A E Malkov
- Laboratory of Neurons Systematic Organization, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - O M Rozanova
- Laboratory of Cell Engineering, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - E N Smirnova
- Laboratory of Cell Engineering, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| | - A E Shemyakov
- Theranostics and Nuclear Medicine Laboratory, Institute of Theoretical and Experimental Biophysics RAS, Pushchino, Moscow Region, Russia
| |
Collapse
|
27
|
Wilcox JA, Estrera R, Boire A. The Spectrum of Headache in Leptomeningeal Metastases: A Comprehensive Review with Clinical Management Guidelines. Curr Pain Headache Rep 2023; 27:695-706. [PMID: 37874457 PMCID: PMC10713777 DOI: 10.1007/s11916-023-01180-9] [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] [Accepted: 09/14/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE OF REVIEW Headaches are a common, oftentimes debilitating symptom in patients with leptomeningeal metastases. RECENT FINDINGS The third edition of the International Classification of Headache Disorders provides a useful diagnostic framework for headaches secondary to leptomeningeal metastases based on the temporal relationship of headache with disease onset, change in headache severity in correlation with leptomeningeal disease burden, and accompanying neurologic signs such as cranial nerve palsies and encephalopathy. However, headaches in patients with leptomeningeal metastases can be further defined by a wide range of varying cancer- and treatment-related pathophysiologies, each requiring a tailored approach. A thorough review of the literature and expert opinion on five observed headache sub-classifications in patients with leptomeningeal metastases is provided, with attention to necessary diagnostic testing, recommended first-line treatments, and prevention strategies.
Collapse
Affiliation(s)
- Jessica A Wilcox
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rachel Estrera
- Human Oncology and Pathogenesis Program, Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne Boire
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
28
|
Zhao N, Chung TD, Guo Z, Jamieson JJ, Liang L, Linville RM, Pessell AF, Wang L, Searson PC. The influence of physiological and pathological perturbations on blood-brain barrier function. Front Neurosci 2023; 17:1289894. [PMID: 37937070 PMCID: PMC10626523 DOI: 10.3389/fnins.2023.1289894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
The blood-brain barrier (BBB) is located at the interface between the vascular system and the brain parenchyma, and is responsible for communication with systemic circulation and peripheral tissues. During life, the BBB can be subjected to a wide range of perturbations or stresses that may be endogenous or exogenous, pathological or therapeutic, or intended or unintended. The risk factors for many diseases of the brain are multifactorial and involve perturbations that may occur simultaneously (e.g., two-hit model for Alzheimer's disease) and result in different outcomes. Therefore, it is important to understand the influence of individual perturbations on BBB function in isolation. Here we review the effects of eight perturbations: mechanical forces, temperature, electromagnetic radiation, hypoxia, endogenous factors, exogenous factors, chemical factors, and pathogens. While some perturbations may result in acute or chronic BBB disruption, many are also exploited for diagnostic or therapeutic purposes. The resultant outcome on BBB function depends on the dose (or magnitude) and duration of the perturbation. Homeostasis may be restored by self-repair, for example, via processes such as proliferation of affected cells or angiogenesis to create new vasculature. Transient or sustained BBB dysfunction may result in acute or pathological symptoms, for example, microhemorrhages or hypoperfusion. In more extreme cases, perturbations may lead to cytotoxicity and cell death, for example, through exposure to cytotoxic plaques.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - Tracy D. Chung
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Zhaobin Guo
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
| | - John J. Jamieson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lily Liang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raleigh M. Linville
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alex F. Pessell
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Linus Wang
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
29
|
Moss TT, Stavem K, Aandahl A, Gløersen AS, Grønberg BH, Neumann K, Vedeler CA, Lundqvist C. Case Report: Limbic encephalitis following treatment with durvalumab for small-cell lung cancer. Front Immunol 2023; 14:1278761. [PMID: 37908347 PMCID: PMC10613972 DOI: 10.3389/fimmu.2023.1278761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 09/29/2023] [Indexed: 11/02/2023] Open
Abstract
Background Durvalumab is an immune checkpoint Inhibitor (ICIs) that is used in the treatment of malignant tumors, such as lung cancer and melanoma. ICIs are associated with immune-related adverse events including autoimmune encephalitis, although both paraneoplastic phenomena and ICI treatment may lead to autoimmunity. Case presentation We describe a 72-year old male patient with small-cell lung cancer, who during adjuvant treatment with Durvalumab developed GABABR1 and GAD65 antibodies and both diabetes and autoimmune limbic encephalitis. Because he was followed prospectively as part of a treatment study, we had access to repeated serum samples and cognitive assessments over time prior to developing encephalitis and diabetes, in addition to later assessments. A high titer of GABABR1 antibodies appeared early, while GAD65 antibodies appeared later with a lower titer in parallel with the development of diabetes. As he subsequently developed clinical signs of encephalitis, verified by EEG and brain MRI, he also had CSF GABABR1 antibodies. Durvalumab was discontinued and steroid treatment with subsequent plasmapheresis were started, resulting in reduction of both CSF and serum antibody levels. Clinical signs of encephalitis gradually improved. Conclusion This case illustrates the importance of being aware of possible serious autoimmune adverse reactions, including neurological syndromes such as encephalitis, when treating patients with high risk of para-neoplasia with ICIs. In addition, the case shows the development of autoantibodies over time.
Collapse
Affiliation(s)
- Thomas T. Moss
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Knut Stavem
- Pulmonary Department, Akershus University Hospital, Lørenskog, Norway
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Astrid Aandahl
- Department of Immunology and Transfusion Medicine, Akershus University Hospital, Lørenskog, Norway
| | - Anne S. Gløersen
- Pulmonary Department, Akershus University Hospital, Lørenskog, Norway
| | - Bjørn H. Grønberg
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Oncology, St. Olavs Hospital, Trondheim, Norway
| | - Kirill Neumann
- Pulmonary Department, Akershus University Hospital, Lørenskog, Norway
| | - Christian A. Vedeler
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Christofer Lundqvist
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
- Health Services Research Unit, Akershus University Hospital, Lørenskog, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
30
|
Peterson RK, King TZ. A systematic review of pediatric neuropsychological outcomes with proton versus photon radiation therapy: A call for equity in access to treatment. J Int Neuropsychol Soc 2023; 29:798-811. [PMID: 36323679 DOI: 10.1017/s1355617722000819] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE There is increasing interest in the utilization of proton beam radiation therapy (PRT) to treat pediatric brain tumors based upon presumed advantages over traditional photon radiation therapy (XRT). PRT provides more conformal radiation to the tumor with reduced dose to healthy brain parenchyma. Less radiation exposure to brain tissue beyond the tumor is thought to reduce neuropsychological sequelae. This systematic review aimed to provide an overview of published studies comparing neuropsychological outcomes between PRT and XRT. METHOD PubMed, PsychINFO, Embase, Web of Science, Scopus, and Cochrane were systematically searched for peer-reviewed published studies that compared neuropsychological outcomes between PRT and XRT in pediatric brain tumor patients. RESULTS Eight studies were included. Six of the studies utilized retrospective neuropsychological data; the majority were longitudinal studies (n = 5). XRT was found to result in lower neuropsychological functioning across time. PRT was associated with generally stable neuropsychological functioning across time, with the exception of working memory and processing speed, which showed variable outcomes across studies. However, studies inconsistently included or considered medical and sociodemographic differences between treatment groups, which may have impacted neuropsychological outcomes. CONCLUSIONS Despite methodological limitations, including limited baseline neuropsychological evaluations, temporal variability between radiation treatment and first evaluation or initial and follow-up evaluations, and heterogenous samples, there is emerging evidence of sociodemographic inequities in access to PRT. With more institutions dedicating funding towards PRT, there may be the opportunity to objectively evaluate the neuropsychological benefits of patients matched on medical and sociodemographic variables.
Collapse
Affiliation(s)
- Rachel K Peterson
- Department of Neuropsychology, Kennedy Krieger Institute, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Tricia Z King
- Department of Psychology, Georgia State University, Atlanta, USA
- Neuroscience Institute, Georgia State University, Atlanta, USA
| |
Collapse
|
31
|
Soliman AM, Ghorab WM, Lotfy DM, Karam HM, Ghorab MM, Ramadan LA. Novel iodoquinazolinones bearing sulfonamide moiety as potential antioxidants and neuroprotectors. Sci Rep 2023; 13:15546. [PMID: 37730974 PMCID: PMC10511408 DOI: 10.1038/s41598-023-42239-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
In a search for new antioxidants, a set of new iodoquinazolinone derivatives bearing benzenesulfonamide moiety and variable acetamide pharmacophores 5-17 were designed and synthesized. The structures of the synthesized compounds were confirmed based on spectral data. Compounds 5-17 were screened using in vitro assay for their antioxidant potential and acetylcholinesterase (AChE) inhibitory activity. The 2-(6-iodo-4-oxo-3-(4-sulfamoylphenyl)-3,4-dihydroquinazolin-2-ylthio)-N-(pyrazin-2-yl) acetamide 14 was the most active scaffold with potent AChE inhibitory activity. Compound 14 showed relative safety with a median lethal dose of 300 mg/kg (LD50 = 300 mg/kg), in an acute toxicity study. The possible antioxidant and neuroprotective activities of 14 were evaluated in irradiated mice. Compound 14 possessed in vivo AChE inhibitory activity and was able to modify the brain neurotransmitters. It was able to cause mitigation of gamma radiation-induced oxidative stress verified by the decline in Myeloperoxidase (MPO) and increase of glutathione (GSH) levels. Also, 14 restored the alterations in behavioral tests. Molecular docking of 14 was performed inside MPO and AChE active sites and showed the same binding interactions as that of the co-crystallized ligands considering the binding possibilities and energy scores. These findings would support that 14 could be considered a promising antioxidant with a neuromodulatory effect.
Collapse
Affiliation(s)
- Aiten M Soliman
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Walid M Ghorab
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Dina M Lotfy
- Pharmacology and Toxicology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Heba M Karam
- Pharmacology and Toxicology Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Mostafa M Ghorab
- Drug Chemistry Laboratory, Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Laila A Ramadan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, Cairo, 11829, Egypt
| |
Collapse
|
32
|
Gan C, Li W, Xu J, Pang L, Tang L, Yu S, Li A, Ge H, Huang R, Cheng H. Advances in the study of the molecular biological mechanisms of radiation-induced brain injury. Am J Cancer Res 2023; 13:3275-3299. [PMID: 37693137 PMCID: PMC10492106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
Radiation therapy is one of the most commonly used treatments for head and neck cancers, but it often leads to radiation-induced brain injury. Patients with radiation-induced brain injury have a poorer quality of life, and no effective treatments are available. The pathogenesis of this condition is unknown. This review summarizes the molecular biological mechanism of radiation-induced brain injury and provides research directions for future studies. The molecular mechanisms of radiation-induced brain injury are diverse and complex. Radiation-induced chronic neuroinflammation, destruction of the blood-brain barrier, oxidative stress, neuronal damage, and physiopathological responses caused by specific exosome secretion lead to radiation-induced brain injury.
Collapse
Affiliation(s)
- Chen Gan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Wen Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Jian Xu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lulian Pang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lingxue Tang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Sheng Yu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Anlong Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Han Ge
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Runze Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical UniversityShenzhen, Guangdong, China
| |
Collapse
|
33
|
Xiao M, Li X, Wang L, Lin B, Zhai M, Hull L, Zizzo A, Cui W, Kiang JG. Skin Wound following Irradiation Aggravates Radiation-Induced Brain Injury in a Mouse Model. Int J Mol Sci 2023; 24:10701. [PMID: 37445879 DOI: 10.3390/ijms241310701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Radiation injury- and radiation combined with skin injury-induced inflammatory responses in the mouse brain were evaluated in this study. Female B6D2F1/J mice were subjected to a sham, a skin wound (SW), 9.5 Gy 60Co total-body gamma irradiation (RI), or 9.5 Gy RI combined with a skin puncture wound (RCI). Survival, body weight, and wound healing were tracked for 30 days, and mouse brain samples were collected on day 30 after SW, RI, RCI, and the sham control. Our results showed that RCI caused more severe animal death and body weight loss compared with RI, and skin wound healing was significantly delayed by RCI compared to SW. RCI and RI increased the chemokines Eotaxin, IP-10, MIG, 6Ckine/Exodus2, MCP-5, and TIMP-1 in the brain compared to SW and the sham control mice, and the Western blot results showed that IP-10 and p21 were significantly upregulated in brain cells post-RI or -RCI. RI and RCI activated both astrocytes and endothelial cells in the mouse brain, subsequently inducing blood-brain barrier (BBB) leakage, as shown by the increased ICAM1 and GFAP proteins in the brain and GFAP in the serum. The Doublecortin (DCX) protein, the "gold standard" for measuring neurogenesis, was significantly downregulated by RI and RCI compared with the sham group. Furthermore, RI and RCI decreased the expression of the neural stem cell marker E-cadherin, the intermediate progenitor marker MASH1, the immature neuron cell marker NeuroD1, and the mature neuron cell marker NeuN, indicating neural cell damage in all development stages after RI and RCI. Immunohistochemistry (IHC) staining further confirmed the significant loss of neural cells in RCI. Our data demonstrated that RI and RCI induced brain injury through inflammatory pathways, and RCI exacerbated neural cell damage more than RI.
Collapse
Affiliation(s)
- Mang Xiao
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Xianghong Li
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Li Wang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Bin Lin
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Min Zhai
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Lisa Hull
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Alex Zizzo
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
| | - Wanchang Cui
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Juliann G Kiang
- Scientific Research Department, Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD 20889, USA
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| |
Collapse
|
34
|
Wang J, Zhang J, Wen W, Wang F, Wu M, Chen D, Yu J. Exploring low-dose radiotherapy to overcome radio-immunotherapy resistance. Biochim Biophys Acta Mol Basis Dis 2023:166789. [PMID: 37302425 DOI: 10.1016/j.bbadis.2023.166789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/13/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the current treatment landscape for cancer, yet the response rates of ICIs remain unmet. Synergistic with immunotherapy, low-dose radiotherapy (LDRT) has been demonstrated to activate anti-tumor immunity - a transition from traditional radiation therapy geared toward local radical treatment to a type of immunological adjuvant. As such, studies utilizing LDRT to enhance the efficacy of immunotherapy have been increasing preclinically and clinically. This paper reviews the recent strategies of using LDRT to overcome the resistance of ICIs, as well as providing potential opportunities in cancer treatment. Despite the potential of LDRT in immunotherapy is recognized, the mechanisms behind this form of treatment remain largely elusive. Thus, we reviewed history, mechanisms and challenges associated with this form of treatment, as well as different modes of its application, to provide relatively accurate practice standards for LDRT as a sensitizing treatment when combined with immunotherapy or radio-immunotherapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Jingxin Zhang
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Weitao Wen
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Fei Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Meng Wu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China
| | - Dawei Chen
- Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| | - Jinming Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, PR China; Shandong University Cancer Center, Jinan, Shandong 250117, PR China; Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, PR China; Research Unit of Radiation Oncology, Chinese Academy of Medical Sciences, Jinan, Shandong 250117, PR China.
| |
Collapse
|
35
|
Oyefeso FA, Goldberg G, Opoku NYPS, Vazquez M, Bertucci A, Chen Z, Wang C, Muotri AR, Pecaut MJ. Effects of acute low-moderate dose ionizing radiation to human brain organoids. PLoS One 2023; 18:e0282958. [PMID: 37256873 PMCID: PMC10231836 DOI: 10.1371/journal.pone.0282958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/27/2023] [Indexed: 06/02/2023] Open
Abstract
Human exposure to low-to-moderate dose ionizing radiation (LMD-IR) is increasing via environmental, medical, occupational sources. Acute exposure to LMD-IR can cause subclinical damage to cells, resulting in altered gene expression and cellular function within the human brain. It has been difficult to identify diagnostic and predictive biomarkers of exposure using traditional research models due to factors including lack of 3D structure in monolayer cell cultures, limited ability of animal models to accurately predict human responses, and technical limitations of studying functional human brain tissue. To address this gap, we generated brain/cerebral organoids from human induced pluripotent stem cells to study the radiosensitivity of human brain cells, including neurons, astrocytes, and oligodendrocytes. While organoids have become popular models for studying brain physiology and pathology, there is little evidence to confirm that exposing brain organoids to LMD-IR will recapitulate previous in vitro and in vivo observations. We hypothesized that exposing brain organoids to proton radiation would (1) cause a time- and dose-dependent increase in DNA damage, (2) induce cell type-specific differences in radiosensitivity, and (3) increase expression of oxidative stress and DNA damage response genes. Organoids were exposed to 0.5 or 2 Gy of 250 MeV protons and samples were collected at 30 minute, 24 hour, and 48 hour timepoints. Using immunofluorescence and RNA sequencing, we found time- and dose-dependent increases in DNA damage in irradiated organoids; no changes in cell populations for neurons, oligodendrocytes, and astrocytes by 24 hours; decreased expression of genes related to oligodendrocyte lineage, astrocyte lineage, mitochondrial function, and cell cycle progression by 48 hours; increased expression of genes related to neuron lineage, oxidative stress, and DNA damage checkpoint regulation by 48 hours. Our findings demonstrate the possibility of using organoids to characterize cell-specific radiosensitivity and early radiation-induced gene expression changes within the human brain, providing new avenues for further study of the mechanisms underlying acute neural cell responses to IR exposure at low-to-moderate doses.
Collapse
Affiliation(s)
- Foluwasomi A. Oyefeso
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Gabriela Goldberg
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Nana Yaa P. S. Opoku
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Marcelo Vazquez
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Antonella Bertucci
- Center for Genomics, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Zhong Chen
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Charles Wang
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
- Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Alysson R. Muotri
- Department of Radiation Medicine, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Michael J. Pecaut
- Department of Biomedical Engineering Sciences, School of Medicine, Loma Linda University, Loma Linda, California, United States of America
- Departments of Pediatrics and Cellular & Molecular Medicine, School of Medicine, Center for Academic Research and Training in Anthropogeny (CARTA), Kavli Institute for Brain and Mind, Archealization Center (ArchC), University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
36
|
Yu Y, Lin X, Feng F, Wei Y, Wei S, Gong Y, Guo C, Wang Q, Shuai P, Wang T, Qin H, Li G, Yi L. Gut microbiota and ionizing radiation-induced damage: Is there a link? ENVIRONMENTAL RESEARCH 2023; 229:115947. [PMID: 37080277 DOI: 10.1016/j.envres.2023.115947] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
According to observational findings, ionizing radiation (IR) triggers dysbiosis of the intestinal microbiota, affecting the structural composition, function, and species of the gut microbiome and its metabolites. These modifications can further exacerbate IR-induced damage and amplify proinflammatory immune responses. Conversely, commensal bacteria and favorable metabolites can remodel the IR-disturbed gut microbial structure, promote a balance between anti-inflammatory and proinflammatory mechanisms in the body, and mitigate IR toxicity. The discovery of effective and safe remedies to prevent and treat radiation-induced injuries is vitally needed because of the proliferation of radiation toxicity threats produced by recent radiological public health disasters and increasing medical exposures. This review examines how the gut microbiota and its metabolites are linked to the processes of IR-induced harm. We highlight protective measures based on interventions with gut microbes to optimize the distress caused by IR damage to human health. We offer prospects for research in emerging and promising areas targeting the prevention and treatment of IR-induced damage.
Collapse
Affiliation(s)
- Yueqiu Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Feiyang Feng
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuanyun Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaqi Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Caimao Guo
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qingyu Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Peimeng Shuai
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tiantian Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guoqing Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
37
|
Zhang C, Zheng J, Chen W, Yang W, Tan X, Fan X, Shen G, Qu L, Chen Z, Shi C. Mitochondrial-targeting fluorescent small molecule IR-780 alleviates radiation-induced brain injury. Brain Res 2023; 1805:148285. [PMID: 36801209 DOI: 10.1016/j.brainres.2023.148285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/12/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
Radiation-induced brain injury (RIBI) is a common complication of radiation therapy for brain tumors. Vascular damage is one of the key factors closely related to the severity of the RIBI. However, effective vascular target treatment strategies are lacking. Previously, we have identified a fluorescent small molecule dye, IR-780, which shows the properties of injury tissue targeting and provided protection against various injuries by modulating oxidative stress. This study aims to validate the therapeutic effect of IR-780 on RIBI. The effectiveness of IR-780 against RIBI has been comprehensively evaluated through techniques such as behavior, immunofluorescence staining, quantitative real-time polymerase chain reaction, Evans Blue leakage experiments, electron microscopy, and flow cytometry. Results show that IR-780 improves cognitive dysfunction, reduces neuroinflammation, restores the expression of tight junction proteins in the blood-brain barrier (BBB), and promotes the recovery of BBB function after whole brain irradiation. IR-780 also accumulates in injured cerebral microvascular endothelial cells, and its subcellular location is in the mitochondria. More importantly, IR-780 can reduce the levels of cellular reactive oxygen species and apoptosis. Moreover, IR-780 has no significant toxic side effects. IR-780 alleviates RIBI by protecting vascular endothelial cells from oxidative stress, reducing neuroinflammation, and restoring BBB function, suggesting IR-780 as a promising treatment candidate for RIBI therapy.
Collapse
Affiliation(s)
- Can Zhang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Jiancheng Zheng
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Wanchao Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Wei Yang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China; Department of Oncology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, China
| | - Xu Tan
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Army Medical University, 400038 Chongqing, China
| | - Gufang Shen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Langfan Qu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China
| | - Zelin Chen
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China.
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, 400038 Chongqing, China.
| |
Collapse
|
38
|
The Effects of Galactic Cosmic Rays on the Central Nervous System: From Negative to Unexpectedly Positive Effects That Astronauts May Encounter. BIOLOGY 2023; 12:biology12030400. [PMID: 36979092 PMCID: PMC10044754 DOI: 10.3390/biology12030400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
Galactic cosmic rays (GCR) pose a serious threat to astronauts’ health during deep space missions. The possible functional alterations of the central nervous system (CNS) under GCR exposure can be critical for mission success. Despite the obvious negative effects of ionizing radiation, a number of neutral or even positive effects of GCR irradiation on CNS functions were revealed in ground-based experiments with rodents and primates. This review is focused on the GCR exposure effects on emotional state and cognition, emphasizing positive effects and their potential mechanisms. We integrate these data with GCR effects on adult neurogenesis and pathological protein aggregation, forming a complete picture. We conclude that GCR exposure causes multidirectional effects on cognition, which may be associated with emotional state alterations. However, the irradiation in space-related doses either has no effect or has performance enhancing effects in solving high-level cognition tasks and tasks with a high level of motivation. We suppose the model of neurotransmission changes after irradiation, although the molecular mechanisms of this phenomenon are not fully understood.
Collapse
|
39
|
Radwan RR, Mohamed HA. Mechanistic approach of the therapeutic potential of mesenchymal stem cells on brain damage in irradiated mice: emphasis on anti-inflammatory and anti-apoptotic effects. Int J Radiat Biol 2023; 99:1463-1472. [PMID: 35647928 DOI: 10.1080/09553002.2022.2084170] [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: 09/15/2021] [Revised: 04/04/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Brain damage which has been induced by radiation generally occurs in radiotherapeutics patients. Stem cell transplantation represents a vital applicant for alleviating neurodegenerative disorders. This work aims at exploring the potential of bone marrow-derived mesenchymal stem cells (BM-MSCs) on brain injury induced by γ radiation in mice and the possible underlying mechanisms were elucidated. MATERIALS AND METHODS Mice were allocated into three groups; Group I (Control), Group II (Irradiated control) where mice submitted to 5 Gy of whole-body γ radiation, Group III (Irradiated + BM-MSCs) where mice were intravenously injected of BM-MSCs at a dose of 106 cells/mice 24 h following irradiation. Animals were sacrificed 28 d following exposure to γ radiation. RESULTS It was observed that BM-MSCs therapy provided a valuable tissue repair as evidenced by a reduction in inflammatory mediators including tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), nuclear factor kappa (NF-κβ), phosphorylated NF-κβ-p65 (P-NF-κβ-p65), interferon-gamma (IFNγ) and monocyte chemoattractant protein-1 (MCP-1) associated with decreased levels of transforming growth factor-β (TGF-β) and vascular endothelial growth factor (VEGF) in brain tissues of irradiated mice. Furthermore, neuronal apoptosis was declined in brain tissues of the BM-MSCs group as remarkable inhibition of caspase-3 and Bax accompanied by elevation of Bcl-2 proteins expression. These results were supported by histopathological investigation. CONCLUSIONS In conclusion, BM-MSCs could display a vital rule in alleviating brain injury in radio-therapeutic patients.
Collapse
Affiliation(s)
- Rasha R Radwan
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Egypt
| | - Heba A Mohamed
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Egypt
| |
Collapse
|
40
|
Hardy SJ, Finkelstein AJ, Tivarus M, Culakova E, Mohile N, Weber M, Lin E, Zhong J, Usuki K, Schifitto G, Milano M, Janelsins-Benton MC. Cognitive and neuroimaging outcomes in individuals with benign and low-grade brain tumours receiving radiotherapy: a protocol for a prospective cohort study. BMJ Open 2023; 13:e066458. [PMID: 36792323 PMCID: PMC9933762 DOI: 10.1136/bmjopen-2022-066458] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/27/2023] [Indexed: 02/17/2023] Open
Abstract
INTRODUCTION Radiation-induced cognitive decline (RICD) occurs in 50%-90% of adult patients 6 months post-treatment. In patients with low-grade and benign tumours with long expected survival, this is of paramount importance. Despite advances in radiation therapy (RT) treatment delivery, better understanding of structures important for RICD is necessary to improve cognitive outcomes. We hypothesise that RT may affect network topology and microstructural integrity on MRI prior to any gross anatomical or apparent cognitive changes. In this longitudinal cohort study, we aim to determine the effects of RT on brain structural and functional integrity and cognition. METHODS AND ANALYSIS This study will enroll patients with benign and low-grade brain tumours receiving partial brain radiotherapy. Patients will receive either hypofractionated (>2 Gy/fraction) or conventionally fractionated (1.8-2 Gy/fraction) RT. All participants will be followed for 12 months, with MRIs conducted pre-RT and 6-month and 12 month post-RT, along with a battery of neurocognitive tests and questionnaires. The study was initiated in late 2018 and will continue enrolling through 2024 with final follow-ups completing in 2025. The neurocognitive battery assesses visual and verbal memory, attention, executive function, processing speed and emotional cognition. MRI protocols incorporate diffusion tensor imaging and resting state fMRI to assess structural connectivity and functional connectivity, respectively. We will estimate the association between radiation dose, imaging metrics and cognitive outcomes. ETHICS AND DISSEMINATION This study has been approved by the Research Subjects Review Board at the University of Rochester (STUDY00001512: Cognitive changes in patients receiving partial brain radiation). All results will be published in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER ClinicalTrials.gov NCT04390906.
Collapse
Affiliation(s)
- Sara J Hardy
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Alan J Finkelstein
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Madalina Tivarus
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Eva Culakova
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| | - Nimish Mohile
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Miriam Weber
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward Lin
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Jianhui Zhong
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
- Center for Advanced Brain Imaging and Neurophysiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenneth Usuki
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - Giovanni Schifitto
- Department of Neurology, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, New York, USA
| | - Michael Milano
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
| | - M C Janelsins-Benton
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
41
|
Perez WD, Perez-Torres CJ. Neurocognitive and radiological changes after cranial radiation therapy in humans and rodents: a systematic review. Int J Radiat Biol 2023; 99:119-137. [PMID: 35511499 DOI: 10.1080/09553002.2022.2074167] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Radiation-induced brain injury is a common long-term side effect for brain cancer survivors, leading to a reduced quality of life. Although there is growing research pertaining to this topic, the relationship between cognitive and radiologically detected lesions of radiation-induced brain injury in humans remains unclear. Furthermore, clinically translatable similarities between rodent models and human findings are also undefined. The objective of this review is to then identify the current evidence of radiation-induced brain injury in humans and to compare these findings to current rodent models of radiation-induced brain injury. METHODS This review includes an examination of the current literature on cognitive and radiological characteristics of radiation-induced brain injury in humans and rodents. A thorough search was conducted on PubMed, Web of Science, and Scopus to identify studies that performed cognitive assessments and magnetic resonance imaging techniques on either humans or rodents after cranial radiation therapy. A qualitative synthesis of the data is herein reported. RESULTS A total of 153 studies pertaining to cognitively or radiologically detected radiation injury of the brain are included in this systematic review; 106 studies provided data on humans while 47 studies provided data on rodents. Cognitive deficits in humans manifest across multiple domains after brain irradiation. Radiological evidence in humans highlight various neuroimaging-detectable changes post-irradiation. It is unclear, however, whether these findings reflect ground truth or research interests. Additionally, rodent models do not comprehensively reproduce characteristics of cognitive and radiological injury currently identified in humans. CONCLUSION This systematic review demonstrates that associations between and within cognitive and radiological radiation-induced brain injuries often rely on the type of assessment. Well-designed studies that evaluate the spectrum of potential injury are required for a precise understanding of not only the clinical significance of radiation-induced brain injury in humans, but also how to replicate injury development in pre-clinical models.
Collapse
Affiliation(s)
- Whitney D Perez
- School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Carlos J Perez-Torres
- School of Health Sciences, Purdue University, West Lafayette, IN, USA.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, USA.,Academy of Integrated Science, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| |
Collapse
|
42
|
Unraveling the peripheral and local role of inflammatory cytokines in glioblastoma survival. Cytokine 2023; 161:156059. [PMID: 36272241 DOI: 10.1016/j.cyto.2022.156059] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 09/22/2022] [Accepted: 09/24/2022] [Indexed: 11/07/2022]
Abstract
Glioblastoma (GBM) is a life-threatening disease that presents high morbidity and mortality. The standardized treatment protocol results in a global survival of less than three years in the majority of cases. Immunotherapies have gained wide recognition in cancer treatment; however, GBM has an immunosuppressive microenvironment diminishing the possible effectiveness of this therapy. In this sense, investigating the inflammatory settings and the tumoral nature of GBM patients are an important goal to create an individual plan of treatment to improve overall survival rate and quality of life of these patients. Thirty-two patients who underwent surgical resection of GBM were included in this study. Tumor samples and 10 mL of peripheral blood were collected and immediately frozen. TNF-a, IL-1a and IL-4 were evaluated in the tumor and TNF-a, IL-1a and TGF-b in the plasma by Luminex assay. Immunohistochemistry analysis to determine immune celular profile was done, including immunohistochemistry for CD20, CD68 and CD3. Three cases were excluded. Tumor topography, tumor nature, and tumor volume reconstructions were accurately analyzed by T1-weighted, T2-weighted, and FLAIR magnetic resonance imaging. We found that GBM patients with below median peripheral levels of TNF-a and IL-1a had a decreased survival rate when compared to above median patients. On the other hand, patients with below median peripheral levels of TGF-b increased overall survival rate. Intratumoral IL-1a above median was associated with higher number of macrophages and fewer with B cells. Furthermore, plasmatic TNF-a levels were correlated with intratumoral TNF-a levels, suggesting that peripheral cytokines are related to the tumoral microenvironment. Even though tumor size has no difference regarding survival rate, we found a negative correlation between intratumoral IL-4 and tumor size, where larger tumors have less IL-4 expression. Nevertheless, the tumoral nature had a significant effect in overall survival rate, considering that infiltrative tumors showed decreased survival rate and intratumoral TNF-a. Moreover, expansive tumors revealed fewer macrophages and higher T cells. In multiple variation analyzes, we demonstrated that infiltrative tumors and below median peripheral IL-1a expression represent 3 times and 5 times hazard ratio, respectively, demonstrating a poor prognosis. Here we found that peripheral cytokines had a critical role as prognostic tools in a small cohort of GBM patients.
Collapse
|
43
|
Krauss P, Steininger K, Motov S, Sommer B, Bonk MN, Cortes A, Wolfert C, Stueben G, Shiban E, Kahl KH. Resection of supratentorial brain metastases with intraoperative radiotherapy. Is it safe? Analysis and experiences of a single center cohort. Front Surg 2022; 9:1071804. [PMID: 36632525 PMCID: PMC9826792 DOI: 10.3389/fsurg.2022.1071804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/12/2022] [Indexed: 12/27/2022] Open
Abstract
Introduction Intraoperative Radiotherapy (ioRT) is an emerging treatment option in oncologic surgery for various diseases including intraaxial brain lesions to improve surgical outcome and accelerate the adjuvant oncologic therapy. Despite its use in glioma surgery, the application and data regarding ioRT in the treatment of brain metastases (BMs) is sparse. Here were report the largest series of supratentorial BMs treated with resection and ioRT according to functional outcome and adverse events. Methods We performed a retrospective chart review analysis of patients undergoing surgery for BMs following an interdisciplinary tumor board decision in every case with ioRT at our institution. Patient properties, functional status (Karnofsky Performance Score/KPS) before and after surgery as well as oncologic (disease, recursive partitioning analysis, lesion size) and operative parameters were analyzed until hospital discharge. Adverse events (AE) were recorded until 30 days after surgery and rated according to the Clavien Dindo Grading (CDG) scale. Results 70 patients (40 female) with various oncologic diseases were identified and analyzed. Six underwent prior RT. Mean age was 66 ± 11 years. Preoperative median KPS was 80% with a mean BM volume of 3.2 ± 1.2 cm3. Nine patients (13%) experienced in total 14 AEs, including 2 cases (3%) of postoperative death (CDG5) and 2 with new postoperative epilepsy necessitating additional pharmacotreatment (CDG2). Five patients suffered from new neurologic deficit (CDG1) not needing further surgical or medical treatment. After surgery, the neurological status in 7 patients (10%) deteriorated while it improved in 21 cases (30%). Patients experiencing AEs had longer hospitalization and poorer postoperative KPS mdn. 90 vs. 80%. There was no statistically significant deterioration of the functional status during the immediate postoperative course in the whole patient cohort. Conclusion Surgery for supratentorial BMs with ioRT seems safe and feasible. Further studies on the benefit regarding oncologic outcome need to be performed.
Collapse
Affiliation(s)
- Philipp Krauss
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany,Correspondence: Philipp Krauss
| | - Kathrin Steininger
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | - Stefan Motov
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | - Bjoern Sommer
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | | | - Abraham Cortes
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | - Christina Wolfert
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | - Georg Stueben
- Department of Radiooncology, University Hospital Augsburg, Augsburg, Germany
| | - Ehab Shiban
- Department of Neurosurgery, University Hospital Augsburg, Augsburg, Germany
| | - Klaus Henning Kahl
- Department of Radiooncology, University Hospital Augsburg, Augsburg, Germany
| |
Collapse
|
44
|
Novel Therapeutic Approaches in Neoplastic Meningitis. Cancers (Basel) 2022; 15:cancers15010119. [PMID: 36612116 PMCID: PMC9817816 DOI: 10.3390/cancers15010119] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/18/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Central nervous system (CNS) metastasis from systemic cancers can involve the brain parenchyma, leptomeninges, or the dura. Neoplastic meningitis (NM), also known by different terms, including leptomeningeal carcinomatosis and carcinomatous meningitis, occurs due to solid tumors and hematologic malignancies and is associated with a poor prognosis. The current management paradigm entails a multimodal approach focused on palliation with surgery, radiation, and chemotherapy, which may be administered systemically or directly into the cerebrospinal fluid (CSF). This review focuses on novel therapeutic approaches, including targeted and immunotherapeutic agents under investigation, that have shown promise in NM arising from solid tumors.
Collapse
|
45
|
Iturri L, Bertho A, Lamirault C, Juchaux M, Gilbert C, Espenon J, Sebrie C, Jourdain L, Pouzoulet F, Verrelle P, De Marzi L, Prezado Y. Proton FLASH Radiation Therapy and Immune Infiltration: Evaluation in an Orthotopic Glioma Rat Model. Int J Radiat Oncol Biol Phys 2022:S0360-3016(22)03639-2. [PMID: 36563907 DOI: 10.1016/j.ijrobp.2022.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 12/02/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE FLASH radiation therapy (FLASH-RT) is a promising radiation technique that uses ultrahigh doses of radiation to increase the therapeutic window of the treatment. FLASH-RT has been observed to provide normal tissue sparing at high dose rates and similar tumor control compared with conventional RT, yet the biological processes governing these radiobiological effects are still unknown. In this study, we sought to investigate the potential immune response generated by FLASH-RT in a high dose of proton therapy in an orthotopic glioma rat model. METHODS AND MATERIALS We cranially irradiated rats with a single high dose (25 Gy) using FLASH dose rate proton irradiation (257 ± 2 Gy/s) or conventional dose rate proton irradiation (4 ± 0.02 Gy/s). We first assessed the protective FLASH effect that resulted in our setup through behavioral studies in naïve rats. This was followed by a comprehensive analysis of immune cells in blood, healthy tissue of the brain, and tumor microenvironment by flow cytometry. RESULTS Proton FLASH-RT spared memory impairment produced by conventional high-dose proton therapy and induced a similar tumor infiltrating lymphocyte recruitment. Additionally, a general neuroinflammation that was similar in both dose rates was observed. CONCLUSIONS Overall, this study demonstrated that FLASH proton therapy offers a neuro-protective effect even at high doses while mounting an effective lymphoid immune response in the tumor.
Collapse
Affiliation(s)
- Lorea Iturri
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France.
| | - Annaïg Bertho
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Charlotte Lamirault
- Institut Curie, PSL University, Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiotherapy (RadeXp), Paris, France
| | - Marjorie Juchaux
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Cristèle Gilbert
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Julie Espenon
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Catherine Sebrie
- CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, BIOMAPS Université Paris-Saclay, Orsay, France
| | - Laurène Jourdain
- CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, BIOMAPS Université Paris-Saclay, Orsay, France
| | - Frédéric Pouzoulet
- Institut Curie, PSL University, Département de Recherche Translationnelle, CurieCoreTech-Experimental Radiotherapy (RadeXp), Paris, France
| | - Pierre Verrelle
- Institut Curie, Campus Universitaire, PSL Research University, University Paris Saclay, INSERM LITO (U1288), Orsay, 91898 France; Centre de Protonthérapie d'Orsay, Radiation Oncology Department, Campus Universitaire, Institut Curie, PSL Research University, Orsay, 91898 France
| | - Ludovic De Marzi
- Institut Curie, Campus Universitaire, PSL Research University, University Paris Saclay, INSERM LITO (U1288), Orsay, 91898 France; Centre de Protonthérapie d'Orsay, Radiation Oncology Department, Campus Universitaire, Institut Curie, PSL Research University, Orsay, 91898 France
| | - Yolanda Prezado
- Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| |
Collapse
|
46
|
Sharma S, Borski C, Hanson J, Garcia MA, Link CD, Hoeffer C, Chatterjee A, Nagpal P. Identifying an Optimal Neuroinflammation Treatment Using a Nanoligomer Discovery Engine. ACS Chem Neurosci 2022; 13:3247-3256. [PMID: 36410860 DOI: 10.1021/acschemneuro.2c00365] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Acute activation of innate immune response in the brain, or neuroinflammation, protects this vital organ from a range of external pathogens and promotes healing after traumatic brain injury. However, chronic neuroinflammation leading to the activation of immune cells like microglia and astrocytes causes damage to the nervous tissue, and it is causally linked to a range of neurodegenerative diseases such as Alzheimer's diseases (AD), Multiple Sclerosis (MS), Parkinson's disease (PD), and many others. While neuroinflammation is a key target for a range of neuropathological diseases, there is a lack of effective countermeasures to tackle it, and existing experimental therapies require fairly invasive intracerebral and intrathecal delivery due to difficulty associated with the therapeutic crossover between the blood-brain barrier, making such treatments impractical to treat neuroinflammation long-term. Here, we present the development of an optimal neurotherapeutic using our Nanoligomer Discovery Engine, by screening downregulation of several proinflammatory cytokines (e.g., Interleukin-1β or IL-1β, tumor necrosis factor-alpha or TNF-α, TNF receptor 1 or TNFR1, Interleukin 6 or IL-6), inflammasomes (e.g., NLRP1), key transcription factors (e.g., nuclear factor kappa-B or NF-κβ) and their combinations, as upstream regulators and canonical pathway targets, to identify and validate the best-in-class treatment. Using our high-throughput drug discovery, target validation, and lead molecule identification via a bioinformatics and artificial intelligence-based ranking method to design sequence-specific peptide molecules to up- or downregulate gene expression of the targeted gene at will, we used our discovery engine to perturb and identify most effective upstream regulators and canonical pathways for therapeutic intervention to reverse neuroinflammation. The lead neurotherapeutic was a combination of Nanoligomers targeted to NF-κβ (SB.201.17D.8_NF-κβ1) and TNFR1 (SB.201.18D.6_TNFR1), which were identified using in vitro cell-based screening in donor-derived human astrocytes and further validated in vivo using a mouse model of lipopolysaccharide (LPS)-induced neuroinflammation. The combination treatment SB_NI_111 was delivered without any special formulation using a simple intraperitoneal injection of low dose (5 mg/kg) and was found to significantly suppress the expression of LPS-induced neuroinflammation in mouse hippocampus. These results point to the broader applicability of this approach towards the development of therapies for chronic neuroinflammation-linked neurodegenerative diseases, sleep countermeasures, and others, and the potential for further investigation of the lead neurotherapeutic molecule as reversible gene therapy.
Collapse
Affiliation(s)
- Sadhana Sharma
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| | - Curtis Borski
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Jessica Hanson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Micklaus A Garcia
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Christopher D Link
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Charles Hoeffer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado 80309, United States.,Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, Colorado 80303, United States
| | - Anushree Chatterjee
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| | - Prashant Nagpal
- Sachi Bioworks, Colorado Technology Center, 685 S Arthur AvenueLouisville, Colorado 80027, United States
| |
Collapse
|
47
|
Gómez-Alonso I, Baltierra-Uribe S, Sánchez-Torres L, Cancino-Diaz M, Cancino-Diaz J, Rodriguez-Martinez S, Ovruski SM, Hendrichs J, Cancino J. Irradiation and parasitism affect the ability of larval hemocytes of Anastrepha obliqua for phagocytosis and the production of reactive oxygen species. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 111:e21953. [PMID: 35927971 DOI: 10.1002/arch.21953] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/07/2022] [Accepted: 07/11/2022] [Indexed: 06/15/2023]
Abstract
The development of the parasitoid Doryctobracon crawfordi (Viereck) (Hymenoptera: Braconidae) in Anastrepha obliqua (McQuart) (Diptera: Tephritidae) larvae is unviable in nature; however, if the host larva is irradiated at 160 Gy, the parasitoid develops and emerges successfully. This suggests that radiation affects the immune responses of A. obliqua larvae, while the underlying mechanisms remain to be revealed. Using optical and electronic microscopies we determined the number and type of hemocyte populations found inside the A. obliqua larvae, either nonirradiated, irradiated at 160 Gy, parasitized by D. crawfordi, or irradiated and parasitized. Based on flow cytometry, the capacity to produce reactive oxygen species (ROS) was determined by the 123-dihydrorhodamine method in those hemocyte cells. Five cell populations were found in the hemolymph of A. obliqua larvae, two of which (granulocytes and plasmatocytes) can phagocytize and produce ROS. A reduction in the number of cells, mainly of the phagocytic type, was observed, as well as the capacity of these cells to produce ROS, when A. obliqua larvae were irradiated. Both radiation and parasitization decreased the ROS production, and when A. obliqua larvae were irradiated followed by parasitization by D. crawfordi, the reduction of the ROS level was even greater. In contrast, a slight increase in the size of these cells was observed in the hemolymph of the parasitized larvae compared to those in nonparasitized larvae. These results suggest that radiation significantly affects the phagocytic cells of A. obliqua and thus permits the development of the parasitoid D. crawfordi.
Collapse
Affiliation(s)
- Itzia Gómez-Alonso
- Posgrado en Ciencias Químico-Biológicas, Instituto Politécnico Nacional, Prolongación del Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Shantal Baltierra-Uribe
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Luvia Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Mario Cancino-Diaz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Juan Cancino-Diaz
- Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Sandra Rodriguez-Martinez
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Prolongación de Carpio y Calle Plan de Ayala s/n, Santo Tomás, Miguel Hidalgo, Mexico City, México
| | - Sergio M Ovruski
- LIEMEN, División Control Biológico de Plagas, PROIMI Biotecnología, CONICET, Ave. Belgrano y Pje. Caseros, San Miguel de Tucumán, Argentina
| | - Jorge Hendrichs
- Division of Nuclear Insect Pest Control Section, Joint FAO/IAEA Techniques in Food and Agriculture, IAEA Wagramerstrasse 5, Vienna, Austria
| | - Jorge Cancino
- Departamento de Control Biológico, Programa Moscafrut SADER-IICA, Camino a Cacahoatales S. N., Metapa de Domínguez, Chiapas, México
| |
Collapse
|
48
|
Moskaleva EY, Rodina AV, Semochkina YP, Vysotskaya OV. Analysis of Markers of Oxidative Damage of Neurons and Neuroinflammation in the Long-Term Period after Gamma Irradiation of a Mouse Head at Different Doses. BIOL BULL+ 2022. [DOI: 10.1134/s1062359022120159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
|
49
|
Rubinstein L, Kiffer F, Puukila S, Lowe MG, Goo B, Luthens A, Schreurs AS, Torres SM, Steczina S, Tahimic CGT, Allen AR. Mitochondria-Targeted Human Catalase in the Mouse Longevity MCAT Model Mitigates Head-Tilt Bedrest-Induced Neuro-Inflammation in the Hippocampus. Life (Basel) 2022; 12:1838. [PMID: 36362993 PMCID: PMC9695374 DOI: 10.3390/life12111838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/28/2022] [Accepted: 11/03/2022] [Indexed: 10/22/2024] Open
Abstract
Microgravity (modeled by head-tilt bedrest and hind-limb unloading), experienced during prolonged spaceflight, results in neurological consequences, central nervous system (CNS) dysfunction, and potentially impairment during the performance of critical tasks. Similar pathologies are observed in bedrest, sedentary lifestyle, and muscle disuse on Earth. In our previous study, we saw that head-tilt bedrest together with social isolation upregulated the milieu of pro-inflammatory cytokines in the hippocampus and plasma. These changes were mitigated in a MCAT mouse model overexpressing human catalase in the mitochondria, pointing out the importance of ROS signaling in this stress response. Here, we used a head-tilt model in socially housed mice to tease out the effects of head-tilt bedrest without isolation. In order to find the underlying molecular mechanisms that provoked the cytokine response, we measured CD68, an indicator of microglial activation in the hippocampus, as well as changes in normal in-cage behavior. We hypothesized that hindlimb unloading (HU) will elicit microglial hippocampal activations, which will be mitigated in the MCAT ROS-quenching mice model. Indeed, we saw an elevation of the activated microglia CD68 marker following HU in the hippocampus, and this pathology was mitigated in MCAT mice. Additionally, we identified cytokines in the hippocampus, which had significant positive correlations with CD68 and negative correlations with exploratory behaviors, indicating a link between neuroinflammation and behavioral consequences. Unveiling a correlation between molecular and behavioral changes could reveal a biomarker indicative of these responses and could also result in a potential target for the treatment and prevention of cognitive changes following long space missions and/or muscle disuse on Earth.
Collapse
Affiliation(s)
- Linda Rubinstein
- Universities Space Research Association USRA, Columbia, MD 21046, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- The Joseph Sagol Neuroscience Center, Sheba Research Hospital, Ramat Gan 52621, Israel
| | - Frederico Kiffer
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stephanie Puukila
- Universities Space Research Association USRA, Columbia, MD 21046, USA
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | - Moniece G Lowe
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Brie Goo
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | | | - Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
| | - Samantha M Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Sonette Steczina
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- Blue Marble Space Institute of Science, Seattle, WA 98154, USA
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, Mountain View, CA 94035, USA
- KBR, Houston, TX 77002, USA
- Department of Biology, University of North Florida, Jacksonville, FL 32224, USA
| | - Antiño R Allen
- Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Radiation Health Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
50
|
Li Q, Zhang Y, Hu J, Yuan B, Zhang P, Wang Y, Jin X, Du L, Jin Y. The Improved Brain-Targeted Drug Delivery of Edaravone Temperature-Sensitive Gels by Ultrasound for γ-ray Radiation-Induced Brain Injury. Pharmaceutics 2022; 14:2281. [PMID: 36365100 PMCID: PMC9698875 DOI: 10.3390/pharmaceutics14112281] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 08/30/2023] Open
Abstract
Radiation-induced brain injury (RBI) is a common neurological disease caused by ionizing radiation (IR). Edaravone (EDA) is a free radical scavenger, has the potential to treat RBI. EDA loaded temperature-sensitive gels (TSGs) were prepared for subcutaneous injection to improve inconvenient administration of intravenous infusion. RBI mice model was established by irradiation of 60Co γ-ray on head. EDA TSGs could improve spontaneous behavior, learning and memory and anxiety of RBI mice by behavior tests, including the open field test, the novel object recognition test, the elevated plus maze test and the fear conditioning test. The therapeutic effects were enhanced with the assistance of ultrasound. Alleviative pathological changes, decreased the expression of Molondialdehyde (MDA) and Interleukin-6 (IL-6) in the hippocampus of brain, indicated reduced oxidative stress and inflammatory response with the treatment of EDA TSGs and ultrasound. Moreover, ultrasound was superior to the use of EDA TSGs. Safe and effective EDA TSGs were prepared for RBI, and the feasibility of brain-targeted drug delivery enhanced by ultrasound was preliminarily demonstrated in this study.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yizhi Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Jinglu Hu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Pengcheng Zhang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Yaxin Wang
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xu Jin
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Lina Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
- School of Pharmacy, Henan University, Kaifeng 475004, China
| |
Collapse
|