1
|
Ding Y, Jiang C, Chen L, Liu X, Shao B. Astragaloside IV confers neuroprotection against radiation-induced neuronal senescence via the ERK pathway. Exp Neurol 2024; 386:115135. [PMID: 39746463 DOI: 10.1016/j.expneurol.2024.115135] [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: 09/13/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Various factors and mechanisms, including radiation, initiate cellular senescence and are concurrent with the progression of various neurodegenerative diseases. Radiation-induced chromosomal aberrations and DNA integrity damage impact the processes of cellular growth, maturation, and aging. Astragaloside IV (AS-IV) has been documented to display significant neuroprotective effects on inflammation, oxidative stress, and cellular apoptosis; however, the precise neuroprotective mechanism of AS-IV against neuronal aging remains unclear. In this study, radiation-induced senescence models in C57BL/6 mice, PC12 cells, and primary neuronal cells were established. SA-β-gal histochemistry, flow cytometric analysis, immunofluorescence technique, and Western blotting analysis were employed to investigate the underlying mechanism of AS-IV in mitigating the aging of the brain cells caused by exposure to radiation. Our findings revealed that radiation exposure may activate the ERK pathway, leading to an increase in SA-β-gal-positive cells, elevated p21 levels, and the arrest of neuronal cells in the G1/S phase. However, AS-IV has been observed to mitigate the radiation-driven proliferation of senescent cells, by downregulating p-ERK and CDK2 expression and upregulating p21 and RB expression in treatment, thereby alleviating the aging and cognitive impairment caused by radiation. Additionally, evidence of U0126 treatment further supports these findings. In summary, our study showed that AS-IV could protect mice from radiation-induced cognitive impairment and reduce cellular senescence by regulating the ERK pathway.
Collapse
Affiliation(s)
- Yanping Ding
- School of Life Sciences, Northwest Normal University, Lanzhou 730070, Gansu Province, China
| | - Chenxin Jiang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Lili Chen
- School of Life Sciences, Northwest Normal University, Lanzhou 730070, Gansu Province, China
| | - Xin Liu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Baoping Shao
- School of Life Sciences, Lanzhou University, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
2
|
Nomura M, Murad NF, Madhavan SS, Mu WC, Eap B, Garcia TY, Aguirre CG, Verdin E, Ellerby L, Furman D, Newman JC. Ketogenic Diet Reduces Age-Induced Chronic Neuroinflammation in Mice. AGING BIOLOGY 2024; 2:20240038. [PMID: 39697898 PMCID: PMC11654834 DOI: 10.59368/agingbio.20240038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The ketone body beta-hydroxybutyrate (BHB) is an acidic energy metabolite that is synthesized during periods of fasting or exercise. Our previous study demonstrated that an every other week cyclic ketogenic diet (Cyclic KD), which induces blood BHB levels similar to those observed during fasting, reduces midlife mortality and improves memory in aging mice. In addition to its canonical role as an energy metabolite, BHB regulates gene expression and inflammatory activation through non-energetic signaling pathways. The precise mechanisms by which BHB or KD affects brain function during aging remain incompletely understood. Using bulk RNA-sequencing (RNA-Seq), we examined whole brain gene expression of 12-month-old C57BL/6JN male mice fed KD for either one week or 14 months. While one-week KD increases some inflammatory gene expression, the 14-month Cyclic KD largely reduces age-induced neuroinflammatory gene expression. Next, a gene expression analysis of human primary brain cells (microglia, astrocytes, and neurons) using RNA-Seq revealed that BHB alone induces a mild level of inflammation in all three cell types. However, BHB inhibits the more pronounced inflammatory gene expression induced by lipopolysaccharide (LPS) in microglia. BHB exhibits a comparable inhibitory effect on LPS-induced inflammation in mouse primary microglia, which we used as an in vitro model to test and exclude known mechanisms by which BHB regulates inflammation and gene expression as responsible for this modulation of LPS-induced inflammatory gene expression. An acidic milieu resulting from BHB may be required for or contribute to the effect. Overall, we observe that BHB has the potential to attenuate the microglial response to inflammatory stimuli, such as LPS. This may contribute to an observed reduction in chronic inflammation in the brain following long-term Cyclic KD treatment in aging mice.
Collapse
Affiliation(s)
| | | | - Sidharth S. Madhavan
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wei-Chieh Mu
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Brenda Eap
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | | | - Carlos Galicia Aguirre
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eric Verdin
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Lisa Ellerby
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
- Stanford 1000 Immunomes Project, Stanford University School of Medicine, Stanford, CA, USA
- Instituto de Investigaciones en Medicina Traslacional, Universidad Austral, Consejo Nacional de Investigaciones Científicas y Técnicas, 1629, Pilar, Argentina
| | - John C. Newman
- Buck Institute for Research on Aging, Novato, CA, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Division of Geriatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
3
|
Dehghan N, Mousavikia SN, Qasempour Y, Azimian H. Radiation-induced senescence in glioblastoma: An overview of the mechanisms and eradication strategies. Life Sci 2024; 359:123218. [PMID: 39510171 DOI: 10.1016/j.lfs.2024.123218] [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: 08/22/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
Radiotherapy as a treatment method for glioblastoma is limited due to the intrinsic apoptosis resistance mechanisms of the tumor. Administration of higher radiation doses contributes to toxicities in normal tissues and organs at risk, like optic chiasma. Cellular senescence represents an alternative mechanism to apoptosis following radiotherapy in glioblastoma, occurring in both normal and neoplastic cells. Although it impedes the growth of tumors and sustains cells in their cycle, it can also act as a cause of tumor development and recurrence following treatment. In this review, we discuss detailed insights into the significance of radiation-induced senescence in glioblastoma and the underlying mechanisms that lead to radioresistance. We also discuss senescence biomarkers and the role of senescence-associated secretory phenotype (SASP) in tumor recurrence. Finally, we review the studies that have administered potential interventions to eradicate or inhibit senescent cells in glioblastoma after treatment with radiation.
Collapse
Affiliation(s)
- Neda Dehghan
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Younes Qasempour
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Wu J, Lv T, Liu Y, Liu Y, Han Y, Liu X, Peng X, Tang F, Cai J. The role of quercetin in NLRP3-associated inflammation. Inflammopharmacology 2024; 32:3585-3610. [PMID: 39306817 DOI: 10.1007/s10787-024-01566-0] [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: 07/05/2024] [Accepted: 08/30/2024] [Indexed: 11/10/2024]
Abstract
Quercetin is a natural flavonoid that is widely found in fruits and vegetables. As an important flavonoid, it exhibits a wide range of biological activities, including antioxidant, anti-inflammatory, antiviral, immunomodulatory, and analgesic activities. Quercetin exerts powerful antioxidant activity by regulating glutathione, enzyme activity, and the production of reactive oxygen species (ROS). Quercetin exerts powerful anti-inflammatory effects by acting on the Nod-like receptor protein 3 (NLRP3) inflammasome. In diabetes, quercetin has been shown to improve insulin sensitivity and reduce high blood sugar level, while, in neurological diseases, it potentially prevents neuronal degeneration and cognitive decline by regulating neuroinflammation. In addition, in liver diseases, quercetin may improve liver inflammation and fibrosis by regulating the NLRP3 activity. In addition, quercetin may improve inflammation in other diseases based on the NLRP3 inflammasome. With this background, in this review, we have discussed the progress in the study on the mechanism of quercetin toward improving inflammation via NLRP3 inflammasome in the past decade. In addition, from the perspective of quercetin glycoside derivatives, the anti-inflammatory mechanism of hyperoside, rutin, and isoquercetin based on NLRP3 inflammasome has been discussed. Moreover, we have discussed the pharmacokinetics of quercetin and its nanoformulation application, with the aim to provide new ideas for further research on the anti-inflammatory effect of quercetin and its glycoside derivatives based on NLRP3 inflammasome, as well as in drug development and application.
Collapse
Affiliation(s)
- Jiaqi Wu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Yu Liu
- Department of Oncology, Gong'an County People's Hospital, Jingzhou, 434000, China
| | - Yifan Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Department of Oncology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, 434023, China
| | - Yukun Han
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Department of Medical Imaging, School of Medicine, and Positron Emission Computed Tomography (PET) Center of the First Affiliated Hospital, Yangtze University, Jingzhou, 434023, China
| | - Xin Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China.
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, 434023, China.
| | - Fengru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, 1 CREATE Way #04-01, CREATE Tower, Singapore, 138602, Singapore.
| | - Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, 434023, China.
| |
Collapse
|
5
|
Mao XW, Pecaut MJ, Stanbouly S, Nelson G. Oxidative stress, neuroinflammation, and the blood-brain barrier biomarkers on the brain response to spaceflight. LIFE SCIENCES IN SPACE RESEARCH 2024; 43:22-28. [PMID: 39521489 DOI: 10.1016/j.lssr.2024.08.001] [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: 06/18/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 11/16/2024]
Abstract
Prolonged spaceflight can induce physiologic and pathologic abnormalities in the central nervous system (CNS). Our knowledge of the adaptive and/or detrimental effects of spaceflight on the structure and function of the nervous system is limited. Substantial effort has been devoted to identifying and developing reliable indicators to characterize and predict CNS injury and dysfunction associated with prolonged exposure to major components of the space environment including microgravity, physiological/psychological stress, and radiation from galactic cosmic rays (GCR) and solar particle events (SPEs) outside of low earth orbit (LEO). The blood-brain barrier (BBB) is a semi-permeable membrane that is essential to maintain homeostasis of the brain microenvironment. Oxidative stress or other environmental stressors may disrupt BBB integrity and increase permeability leading to immune cell infiltration and undesirable neuroinflammation. The focus of this review article is on BBB damage associated with spaceflight and space radiation in rodent and human studies. We will highlight potential biomarkers for this damage, including site-specific and circulating neuroinflammatory factors, BBB structural and brain parenchyma proteins, and neuroimaging tools for BBB damage evaluation. These knowledge will help to understand the risks associated with space travel and are also critical for novel countermeasure development to mitigate the space flight risk to astronaut performances.
Collapse
Affiliation(s)
- Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, USA.
| | - Michael J Pecaut
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, USA
| | - Seta Stanbouly
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, USA
| | - Gregory Nelson
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA, USA
| |
Collapse
|
6
|
Mishra S, Stany B, Das A, Kanagavel D, Vijayan M. A Comprehensive Review of Membrane Transporters and MicroRNA Regulation in Alzheimer's Disease. Mol Neurobiol 2024; 61:8739-8758. [PMID: 38558361 DOI: 10.1007/s12035-024-04135-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
Alzheimer's disease (AD) is a distressing neurodegenerative condition characterized by the accumulation of amyloid-beta (Aβ) plaques and tau tangles within the brain. The interconnectedness between membrane transporters (SLCs) and microRNAs (miRNAs) in AD pathogenesis has gained increasing attention. This review explores the localization, substrates, and functions of SLC transporters in the brain, emphasizing the roles of transporters for glutamate, glucose, nucleosides, and other essential compounds. The examination delves into the significance of SLCs in AD, their potential for drug development, and the intricate realm of miRNAs, encompassing their transcription, processing, functions, and regulation. MiRNAs have emerged as significant players in AD, including those associated with mitochondria and synapses. Furthermore, this review discusses the intriguing nexus of miRNAs targeting SLC transporters and their potential as therapeutic targets in AD. Finally, the review underscores the interaction between SLC transporters and miRNA regulation within the context of Alzheimer's disease, underscoring the need for further research in this area. This comprehensive review aims to shed light on the complex mechanisms underlying the causation of AD and provides insights into potential therapeutic approaches.
Collapse
Affiliation(s)
- Shatakshi Mishra
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - B Stany
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Anushka Das
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India
| | - Deepankumar Kanagavel
- School of Biosciences and Technology, Department of Biotechnology, VIT University, Vellore, Tamil Nadu, 632014, India.
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th Street, Lubbock, TX, 79430, USA.
| |
Collapse
|
7
|
Li L, Liu X, Han C, Tian L, Wang Y, Han B. Ferroptosis in radiation-induced brain injury: roles and clinical implications. Biomed Eng Online 2024; 23:93. [PMID: 39261942 PMCID: PMC11389269 DOI: 10.1186/s12938-024-01288-y] [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: 12/22/2023] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Radiation-induced brain injury (RBI) presents a significant challenge for patients undergoing radiation therapy for head, neck, and intracranial tumors. This review aims to elucidate the role of ferroptosis in RBI and its therapeutic implications. Specifically, we explore how ferroptosis can enhance the sensitivity of tumor cells to radiation while also examining strategies to mitigate radiation-induced damage to normal brain tissues. By investigating the mechanisms through which radiation increases cellular reactive oxygen species (ROS) and initiates ferroptosis, we aim to develop targeted therapeutic strategies that maximize treatment efficacy and minimize neurotoxicity. The review highlights key regulatory factors in the ferroptosis pathway, including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter system Xc- (System Xc-), nuclear factor erythroid 2-related factor 2 (NRF2), Acyl-CoA synthetase long-chain family member 4 (ACSL4), and others, and their interactions in the context of RBI. Furthermore, we discuss the clinical implications of modulating ferroptosis in radiation therapy, emphasizing the potential for selective induction of ferroptosis in tumor cells and inhibition in healthy cells. The development of advanced diagnostic tools and therapeutic strategies targeting ferroptosis offers a promising avenue for enhancing the safety and efficacy of radiation therapy, underscoring the need for further research in this burgeoning field.
Collapse
Affiliation(s)
- Lifang Li
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Xia Liu
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Chunfeng Han
- Department of Pharmacy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Licheng Tian
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Yongzhi Wang
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Baolin Han
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China.
| |
Collapse
|
8
|
Soraci L, Corsonello A, Paparazzo E, Montesanto A, Piacenza F, Olivieri F, Gambuzza ME, Savedra EV, Marino S, Lattanzio F, Biscetti L. Neuroinflammaging: A Tight Line Between Normal Aging and Age-Related Neurodegenerative Disorders. Aging Dis 2024; 15:1726-1747. [PMID: 38300639 PMCID: PMC11272206 DOI: 10.14336/ad.2023.1001] [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: 08/12/2023] [Accepted: 10/01/2023] [Indexed: 02/02/2024] Open
Abstract
Aging in the healthy brain is characterized by a low-grade, chronic, and sterile inflammatory process known as neuroinflammaging. This condition, mainly consisting in an up-regulation of the inflammatory response at the brain level, contributes to the pathogenesis of age-related neurodegenerative disorders. Development of this proinflammatory state involves the interaction between genetic and environmental factors, able to induce age-related epigenetic modifications. Indeed, the exposure to environmental compounds, drugs, and infections, can contribute to epigenetic modifications of DNA methylome, histone fold proteins, and nucleosome positioning, leading to epigenetic modulation of neuroinflammatory responses. Furthermore, some epigenetic modifiers, which combine and interact during the life course, can contribute to modeling of epigenome dynamics to sustain, or dampen the neuroinflammatory phenotype. The aim of this review is to summarize current knowledge about neuroinflammaging with a particular focus on epigenetic mechanisms underlying the onset and progression of neuroinflammatory cascades in the central nervous system; furthermore, we describe some diagnostic biomarkers that may contribute to increase diagnostic accuracy and help tailor therapeutic strategies in patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, Italian National Research Center of Aging (IRCCS INRCA), Cosenza, Italy.
| | - Andrea Corsonello
- Unit of Geriatric Medicine, Italian National Research Center of Aging (IRCCS INRCA), Cosenza, Italy.
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.
| | - Francesco Piacenza
- Advanced Technology Center for Aging Research, Italian National Research Center of Aging (IRCCS INRCA), IRCCS INRCA, Ancona, Italy.
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy.
- Clinic of Laboratory and Precision Medicine, Italian National Research Center of Aging (IRCCS INRCA), Ancona, Italy.
| | | | | | - Silvia Marino
- IRCCS Centro Neurolesi "Bonino-Pulejo”, Messina, Italy.
| | | | - Leonardo Biscetti
- Section of Neurology, Italian National Research Center on Aging (IRCCS INRCA), Ancona, Italy.
| |
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
|
Kilic Durankus N, Samanci Y, Düzkalir AH, Peker S. Unveiling the Efficacy of Gamma Knife Radiosurgery for Tectal Plate Gliomas. Neurosurgery 2024; 94:780-787. [PMID: 37955438 DOI: 10.1227/neu.0000000000002754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/21/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Tectal plate gliomas (TPGs) are midbrain tumors that grow slowly and have a benign clinical course. Most TPGs are low-grade astrocytomas, but they can encompass various histological tumor types. Gamma Knife radiosurgery (GKRS) is being explored as a potentially safe and effective treatment option for TPGs, although research in this area is limited. This study aims to evaluate GKRS's efficacy and safety in patients with TPG and provide a comprehensive review of existing literature on the topic. METHODS This retrospective, single-center study included 48 patients with consecutive TPG who underwent GKRS between September 2005 and June 2022. Patients diagnosed with TPGs based on radiological or tissue-based criteria and who had a minimum follow-up period of 12 months were eligible for inclusion. The primary end points were local control and the absence of GKRS-associated or tumor-associated mortality and morbidity. RESULTS During a median follow-up of 28.5 months (range, 12-128), the radiological assessment showed tumor control in all cases, with 16.7% achieving a complete response and 68.8% achieving a partial response. Pseudoprogression occurred in 6.2% of cases, with onset ranging from 3 to 8 months. Clinical outcomes revealed no permanent neurological deterioration, with symptoms improving in 14.6% of patients and remaining stable in the others. One patient in the pseudoprogression group experienced transient Parinaud syndrome. One patient died during follow-up because of unrelated causes. The mean survival time after GKRS was 123.7 months. None of the clinical, radiological, or radiosurgical variables showed a correlation with partial/complete response, clinical improvement, or overall survival. CONCLUSION There is limited research available on the management of TPGs, and this study presents the largest patient cohort treated with GKRS, along with a substantial follow-up duration. Despite its limitations, this study demonstrates the efficacy and low-risk profile of GKRS for TPGs.
Collapse
Affiliation(s)
| | - Yavuz Samanci
- Department of Neurosurgery, Koc University School of Medicine, Istanbul , Turkey
- Department of Neurosurgery, Gamma Knife Center, Koc University Hospital, Istanbul , Turkey
| | - Ali Haluk Düzkalir
- Department of Neurosurgery, Gamma Knife Center, Koc University Hospital, Istanbul , Turkey
| | - Selcuk Peker
- Department of Neurosurgery, Koc University School of Medicine, Istanbul , Turkey
- Department of Neurosurgery, Gamma Knife Center, Koc University Hospital, Istanbul , Turkey
| |
Collapse
|
11
|
Ibragimova M, Kussainova A, Aripova A, Bersimbaev R, Bulgakova O. The Molecular Mechanisms in Senescent Cells Induced by Natural Aging and Ionizing Radiation. Cells 2024; 13:550. [PMID: 38534394 PMCID: PMC10969416 DOI: 10.3390/cells13060550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024] Open
Abstract
This review discusses the relationship between cellular senescence and radiation exposure. Given the wide range of ionizing radiation sources encountered by people in professional and medical spheres, as well as the influence of natural background radiation, the question of the effect of radiation on biological processes, particularly on aging processes, remains highly relevant. The parallel relationship between natural and radiation-induced cellular senescence reveals the common aspects underlying these processes. Based on recent scientific data, the key points of the effects of ionizing radiation on cellular processes associated with aging, such as genome instability, mitochondrial dysfunction, altered expression of miRNAs, epigenetic profile, and manifestation of the senescence-associated secretory phenotype (SASP), are discussed. Unraveling the molecular mechanisms of cellular senescence can make a valuable contribution to the understanding of the molecular genetic basis of age-associated diseases in the context of environmental exposure.
Collapse
Affiliation(s)
- Milana Ibragimova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Assiya Kussainova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
- Department of Health Sciences, University of Genova, Via Pastore 1, 16132 Genoa, Italy
| | - Akmaral Aripova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Rakhmetkazhi Bersimbaev
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| | - Olga Bulgakova
- Department of General Biology and Genomics, Institute of Cell Biology and Biotechnology, L.N. Gumilyov Eurasian National University, Astana 010008, Kazakhstan; (M.I.); (A.K.); (A.A.); (R.B.)
| |
Collapse
|
12
|
Ruprecht NA, Singhal S, Schaefer K, Panda O, Sens D, Singhal SK. A Review: Multi-Omics Approach to Studying the Association between Ionizing Radiation Effects on Biological Aging. BIOLOGY 2024; 13:98. [PMID: 38392316 PMCID: PMC10886797 DOI: 10.3390/biology13020098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024]
Abstract
Multi-omics studies have emerged as powerful tools for tailoring individualized responses to various conditions, capitalizing on genome sequencing technologies' increasing affordability and efficiency. This paper delves into the potential of multi-omics in deepening our understanding of biological age, examining the techniques available in light of evolving technology and computational models. The primary objective is to review the relationship between ionizing radiation and biological age, exploring a wide array of functional, physiological, and psychological parameters. This comprehensive review draws upon an extensive range of sources, including peer-reviewed journal articles, government documents, and reputable websites. The literature review spans from fundamental insights into radiation effects to the latest developments in aging research. Ionizing radiation exerts its influence through direct mechanisms, notably single- and double-strand DNA breaks and cross links, along with other critical cellular events. The cumulative impact of DNA damage forms the foundation for the intricate process of natural aging, intersecting with numerous diseases and pivotal biomarkers. Furthermore, there is a resurgence of interest in ionizing radiation research from various organizations and countries, reinvigorating its importance as a key contributor to the study of biological age. Biological age serves as a vital reference point for the monitoring and mitigation of the effects of various stressors, including ionizing radiation. Ionizing radiation emerges as a potent candidate for modeling the separation of biological age from chronological age, offering a promising avenue for tailoring protocols across diverse fields, including the rigorous demands of space exploration.
Collapse
Affiliation(s)
- Nathan A Ruprecht
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sonalika Singhal
- Department of Pathology, University of North Dakota, Grand Forks, ND 58202, USA
| | - Kalli Schaefer
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND 58202, USA
| | - Om Panda
- Department of Public Health, University of California Irvine, Irvine, CA 92697, USA
| | - Donald Sens
- Department of Pathology, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sandeep K Singhal
- Department of Biomedical Engineering, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Pathology, University of North Dakota, Grand Forks, ND 58202, USA
| |
Collapse
|
13
|
Thariat J, Little MP, Zablotska LB, Samson P, O’Banion MK, Leuraud K, Bergom C, Girault G, Azimzadeh O, Bouffler S, Hamada N. Radiotherapy for non-cancer diseases: benefits and long-term risks. Int J Radiat Biol 2024; 100:505-526. [PMID: 38180039 PMCID: PMC11039429 DOI: 10.1080/09553002.2023.2295966] [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: 10/11/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
PURPOSE The discovery of X-rays was followed by a variety of attempts to treat infectious diseases and various other non-cancer diseases with ionizing radiation, in addition to cancer. There has been a recent resurgence of interest in the use of such radiotherapy for non-cancer diseases. Non-cancer diseases for which use of radiotherapy has currently been proposed include refractory ventricular tachycardia, neurodegenerative diseases (e.g. Alzheimer's disease and dementia), and Coronavirus Disease 2019 (COVID-19) pneumonia, all with ongoing clinical studies that deliver radiation doses of 0.5-25 Gy in a single fraction or in multiple daily fractions. In addition to such non-cancer effects, historical indications predominantly used in some countries (e.g. Germany) include osteoarthritis and degenerative diseases of the bones and joints. This narrative review gives an overview of the biological rationale and ongoing preclinical and clinical studies for radiotherapy proposed for various non-cancer diseases, discusses the plausibility of the proposed biological rationale, and considers the long-term radiation risks of cancer and non-cancer diseases. CONCLUSIONS A growing body of evidence has suggested that radiation represents a double-edged sword, not only for cancer, but also for non-cancer diseases. At present, clinical evidence has shown some beneficial effects of radiotherapy for ventricular tachycardia, but there is little or no such evidence of radiotherapy for other newly proposed non-cancer diseases (e.g. Alzheimer's disease, COVID-19 pneumonia). Patients with ventricular tachycardia and COVID-19 pneumonia have thus far been treated with radiotherapy when they are an urgent life threat with no efficient alternative treatment, but some survivors may encounter a paradoxical situation where patients were rescued by radiotherapy but then get harmed by radiotherapy. Further studies are needed to justify the clinical use of radiotherapy for non-cancer diseases, and optimize dose to diseased tissue while minimizing dose to healthy tissue.
Collapse
Affiliation(s)
- Juliette Thariat
- Department of Radiation Oncology, Comprehensive Cancer Centre François Baclesse, Caen, France
- Laboratoire de Physique Corpusculaire IN2P3, ENSICAEN/CNRS UMR 6534, Normandie Université, Caen, France
| | - Mark P. Little
- Radiation Epidemiology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Lydia B. Zablotska
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Pamela Samson
- Department of Radiation Oncology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| | - M. Kerry O’Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Klervi Leuraud
- Research Department on Biological and Health Effects of Ionizing Radiation (SESANE), Institute for Radiological Protection and Nuclear Safety (IRSN), Fontenay-aux-Roses, France
| | - Carmen Bergom
- Department of Radiation Oncology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
- Cardio-Oncology Center of Excellence, Washington University, St. Louis, Missouri, USA
| | - Gilles Girault
- Comprehensive Cancer Centre François Baclesse, Medical Library, Caen, France
| | - Omid Azimzadeh
- Federal Office for Radiation Protection (BfS), Section Radiation Biology, Neuherberg, Germany
| | - Simon Bouffler
- Radiation Protection Sciences Division, UK Health Security Agency (UKHSA), Chilton, Didcot, UK
| | - Nobuyuki Hamada
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Abiko, Chiba, Japan
| |
Collapse
|
14
|
Yin G, Wang Q, Lv T, Liu Y, Peng X, Zeng X, Huang J. The Radioprotective Effect of LBP on Neurogenesis and Cognition after Acute Radiation Exposure. Curr Radiopharm 2024; 17:257-265. [PMID: 38204264 PMCID: PMC11327742 DOI: 10.2174/0118744710274008231220055033] [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: 07/25/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Radiation exposure has been linked to the development of brain damage and cognitive impairment, but the protective effect and mechanism of Lycium barbarum pills (LBP) on radiation-induced neurological damage remains to be clarified. METHODS Behavioral tests and immunohistochemical studies were conducted to evaluate the protective effects of LBP extract (10 g/kg orally daily for 4 weeks) against radiation-induced damage on neurogenesis and cognitive function in Balb/c mice exposed to 5.5 Gy X-ray acute radiation. RESULTS The results showed that the LBP extract significantly improved body weight loss, locomotor activity and spatial learning and memory. Immunohistochemical tests revealed that the LBP extract prevented the loss of proliferating cells, newly generated neurons and interneurons, especially in the subgranular area of the dentate gyrus. CONCLUSION The findings suggest that LBP is a potential neuroprotective drug for mitigating radiation-induced neuropsychological disorders.
Collapse
Affiliation(s)
- Gang Yin
- Department of Neurology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Qinqi Wang
- Department of Internal Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yifan Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xianqin Zeng
- Department of Gynaecology and Obstetrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiangrong Huang
- Department of Integrative Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
15
|
Pathak B, Lange TE, Lampe K, Hollander E, Oria M, Murphy KP, Salomonis N, Sertorio M, Oria M. Development of a Single-Neurosphere Culture to Assess Radiation Toxicity and Pre-Clinical Cancer Combination Therapy Safety. Cancers (Basel) 2023; 15:4916. [PMID: 37894283 PMCID: PMC10605382 DOI: 10.3390/cancers15204916] [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: 09/13/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Radiation therapy (RT) is a crucial treatment modality for central nervous system (CNS) tumors but toxicity to healthy CNS tissues remains a challenge. Additionally, environmental exposure to radiation during nuclear catastrophes or space travel presents a risk of CNS toxicity. However, the underlying mechanisms of radiation-induced CNS toxicity are not fully understood. Neural progenitor cells (NPCs) are highly radiosensitive, resulting in decreased neurogenesis in the hippocampus. This study aimed to characterize a novel platform utilizing rat NPCs cultured as 3D neurospheres (NSps) to screen the safety and efficacy of experimental drugs with and without radiation exposure. The effect of radiation on NSp growth and differentiation was assessed by measuring sphere volume and the expression of neuronal differentiation markers Nestin and GFAP and proliferation marker Ki67. Radiation exposure inhibited NSp growth, decreased proliferation, and increased GFAP expression, indicating astrocytic differentiation. RNA sequencing analysis supported these findings, showing upregulation of Notch, BMP2/4, S100b, and GFAP gene expression during astrogenesis. By recapitulating radiation-induced toxicity and astrocytic differentiation, this single-NSp culture system provides a high-throughput preclinical model for assessing the effects of various radiation modalities and evaluates the safety and efficacy of potential therapeutic interventions in combination with radiation.
Collapse
Affiliation(s)
- Bedika Pathak
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Taylor E. Lange
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA;
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA
| | - Kristin Lampe
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Ella Hollander
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Marina Oria
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
| | - Kendall P. Murphy
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA; (B.P.); (K.L.)
- Department of Orthopedic Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center (CCHMC), Cincinnati, OH 45229, USA;
- Departments of Pediatrics and Bioinformatics, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Mathieu Sertorio
- University of Cincinnati Cancer Center, Cincinnati, OH 45267, USA;
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Marc Oria
- University of Cincinnati Cancer Center, Cincinnati, OH 45267, USA;
- Department of Radiation Oncology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- University of Cincinnati Brain Tumor Center, Cincinnati, OH 45219, USA
| |
Collapse
|
16
|
Wang Y, Ren L, Xu L, Wang J, Zhai J, Zhu G. Radiation Induces Bone Microenvironment Disruption by Activating the STING-TBK1 Pathway. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1316. [PMID: 37512126 PMCID: PMC10386124 DOI: 10.3390/medicina59071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Damage to normal bone tissue following therapeutic irradiation (IR) represents a significant concern, as IR-induced bone microenvironment disruption can cause bone loss and create a more favorable environment for tumor metastases. The aim of the present study was to explore the cellular regulatory mechanism of IR-induced bone microenvironment disruption to effectively prevent radiotherapy-associated adverse effects in the future. Materials and Methods: In this study, a mouse model of local IR was established via local irradiation of the left hind limb of BALB/c mice with 12 Gy X-rays, and an in vitro osteocyte (OCY) model was established by exposing osteocyte-like MLO-Y4 cells to 2, 4, and 8 Gy irradiation to analyze multicellular biological injuries and cellular senescence. Small interfering RNA (siRNA) transfection at the cellular level and a selective antagonist intervention C-176 at the animal level were used to explore the potential role of the stimulator of interferon genes (STING) on IR-induced bone microenvironment disruption. Results: The results showed that 12 Gy local IR induces multicellular dysfunction, manifested as ascension of OCYs exfoliation, activation of osteoclastogenesis, degeneration of osteogenesis and fate conversion of adipogenesis, as well as cellular senescence and altered senescence-associated secretory phenotype (SASP) secretion. Furthermore, the expression of STING was significantly elevated, both in the primary OCYs harvested from locally irradiated mice and in vitro irradiated MLO-Y4 cells, accompanied by the markedly upregulated levels of phosphorylated TANK-binding kinase 1 (P-TBK1), RANKL and sclerostin (SOST). STING-siRNA transfection in vitro restored IR-induced upregulated protein expression of P-TBK1 and RANKL, as well as the mRNA expression levels of inflammatory cytokines, such as IL-1α, IL-6 and NF-κB, accompanied by the alleviation of excessive osteoclastogenesis. Finally, administration of the STING inhibitor C-176 mitigated IR-induced activation of osteoclastogenesis and restraint of osteogenesis, ameliorating the IR-induced biological damage of OCYs, consistent with the inhibition of P-TBK1, RANKL and SOST. Conclusions: The STING-P-TBK1 signaling pathway plays a crucial role in the regulation of the secretion of inflammatory cytokines and osteoclastogenesis potential in IR-induced bone microenvironment disruption. The selective STING antagonist can be used to intervene to block the STING pathway and, thereby, repair IR-induced multicellular biological damage and mitigate the imbalance between osteoclastogenesis and osteoblastgenesis.
Collapse
Affiliation(s)
- Yuyang Wang
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
- Shanghai Municipal Center for Disease Control & Prevention, Shanghai 200051, China
| | - Li Ren
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Linshan Xu
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Jianping Wang
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Jianglong Zhai
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Guoying Zhu
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| |
Collapse
|
17
|
Argenziano M, Di Paola A, Rossi F. Childhood Cancer Survivors: An Overview of the Management of Late Effects. Cancers (Basel) 2023; 15:3150. [PMID: 37370760 DOI: 10.3390/cancers15123150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
The collection of papers in this Special Issue entitled "Frailty in Pediatric and Young Adult Cancer Survivors: from bench to bedside" includes six interesting articles (five reviews and one single-center retrospective longitudinal cohort study) presented by expert researchers in the fields of oncology and pediatrics [...].
Collapse
Affiliation(s)
- Maura Argenziano
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio, 4, 80138 Naples, Italy
| | - Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio, 4, 80138 Naples, Italy
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Via Luigi De Crecchio, 4, 80138 Naples, Italy
| |
Collapse
|
18
|
Sleiman A, Lalanne K, Vianna F, Perrot Y, Richaud M, SenGupta T, Cardot-Martin M, Pedini P, Picard C, Nilsen H, Galas S, Adam-Guillermin C. Targeted Central Nervous System Irradiation with Proton Microbeam Induces Mitochondrial Changes in Caenorhabditis elegans. BIOLOGY 2023; 12:839. [PMID: 37372124 DOI: 10.3390/biology12060839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023]
Abstract
Fifty percent of all patients with cancer worldwide require radiotherapy. In the case of brain tumors, despite the improvement in the precision of radiation delivery with proton therapy, studies have shown structural and functional changes in the brains of treated patients with protons. The molecular pathways involved in generating these effects are not completely understood. In this context, we analyzed the impact of proton exposure in the central nervous system area of Caenorhabditis elegans with a focus on mitochondrial function, which is potentially implicated in the occurrence of radiation-induced damage. To achieve this objective, the nematode C. elegans were micro-irradiated with 220 Gy of protons (4 MeV) in the nerve ring (head region) using the proton microbeam, MIRCOM. Our results show that protons induce mitochondrial dysfunction, characterized by an immediate dose-dependent loss of the mitochondrial membrane potential (ΔΨm) associated with oxidative stress 24 h after irradiation, which is itself characterized by the induction of the antioxidant proteins in the targeted region, observed using SOD-1::GFP and SOD-3::GFP strains. Moreover, we demonstrated a two-fold increase in the mtDNA copy number in the targeted region 24 h after irradiation. In addition, using the GFP::LGG-1 strain, an induction of autophagy in the irradiated region was observed 6 h following the irradiation, which is associated with the up-regulation of the gene expression of pink-1 (PTEN-induced kinase) and pdr-1 (C. elegans parkin homolog). Furthermore, our data showed that micro-irradiation of the nerve ring region did not impact the whole-body oxygen consumption 24 h following the irradiation. These results indicate a global mitochondrial dysfunction in the irradiated region following proton exposure. This provides a better understanding of the molecular pathways involved in radiation-induced side effects and may help in finding new therapies.
Collapse
Affiliation(s)
- Ahmad Sleiman
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LMDN, Cadarache, 13115 Saint-Paul-lez-Durance, France
| | - Kévin Lalanne
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LMDN, Cadarache, 13115 Saint-Paul-lez-Durance, France
| | - François Vianna
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LMDN, Cadarache, 13115 Saint-Paul-lez-Durance, France
| | - Yann Perrot
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LDRI, 92262 Fontenay-aux-Roses, France
| | - Myriam Richaud
- IBMM, University of Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Tanima SenGupta
- Section of Clinical Molecular Biology (EpiGen), Akershus University Hospital, 1478 Lørenskog, Norway
| | - Mikaël Cardot-Martin
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LMDN, Cadarache, 13115 Saint-Paul-lez-Durance, France
| | - Pascal Pedini
- Aix Marseille University, CNRS, EFS, ADES, 13288 Marseille, France
| | | | - Hilde Nilsen
- Section of Clinical Molecular Biology (EpiGen), Akershus University Hospital, 1478 Lørenskog, Norway
- Department of Microbiology, Oslo University Hospital, 0372 Oslo, Norway
| | - Simon Galas
- IBMM, University of Montpellier, CNRS, ENSCM, 34093 Montpellier, France
| | - Christelle Adam-Guillermin
- Institut de Radioprotection et de Sûreté Nucléaire, IRSN, PSE-SANTE/SDOS/LMDN, Cadarache, 13115 Saint-Paul-lez-Durance, France
| |
Collapse
|
19
|
Song C, Yin Y, Qin Y, Li T, Zeng D, Ju T, Duan F, Zhang Y, Lu W. Acanthopanax senticosus extract alleviates radiation-induced learning and memory impairment based on neurotransmitter-gut microbiota communication. CNS Neurosci Ther 2023; 29 Suppl 1:129-145. [PMID: 36971202 PMCID: PMC10314102 DOI: 10.1111/cns.14134] [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: 07/14/2022] [Revised: 12/16/2022] [Accepted: 02/15/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Acanthopanax senticosus (AS) is a medicinal and food plant with many physiological functions, especially nerve protection. Its extract has many functional components, including polysaccharides, flavonoids, saponins, and amino acids. Our previous study indicated that AS extract protected against nerve damage caused by radiation. However, little is known about the gut-brain axis mechanism of AS and its impact on radiation-induced learning and memory impairment. METHOD In 60 Co-γ ray-irradiated mice, we investigated the changes in behavior, neurotransmitters and gut microbiota after different days of administration of AS extract as a dietary supplement. RESULTS The AS extract improved learning and memory ability in mice, and the neurotransmitter levels in the hippocampus and colon started to change from the 7th day, which accompanied changes of the gut microbiota, a decreased abundance of Helicobacter on the 7th day and an increased abundance of Lactobacillus on the 28th day. Among the marker bacteria, Ruminococcus and Clostridiales were associated with 5-HT synthesis, and Streptococcus were associated with 5-HT and ACH synthesis. In addition, the AS extract increased the tight junction protein, inhibited inflammation levels in colon, and even increased the relative protein expression of BDNF and NF-κB and decreased the relative protein expression of IκBα in the hippocampus of irradiated mice. CONCLUSION These results will lay the foundation for further study on the mechanism of the gut-brain axis of AS in preventing radiation-induced learning and memory impairment.
Collapse
Affiliation(s)
- Chen Song
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- School of Chemistry and Chemical EngineeringHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Yishu Yin
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- School of Chemistry and Chemical EngineeringHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Yue Qin
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Tianzhu Li
- ZhenBaoDao Pharmaceutical Co., Ltd150040HarbinChina
| | - Deyong Zeng
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- School of Chemistry and Chemical EngineeringHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Ting Ju
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- School of Chemistry and Chemical EngineeringHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Fangyuan Duan
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- School of Chemistry and Chemical EngineeringHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Yingchun Zhang
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| | - Weihong Lu
- School of Medicine and HealthHarbin Institute of Technology150001HarbinChina
- National and Local Joint Engineering Laboratory for Synthesis, Transformation and Separation of Extreme Environmental NutrientsHarbin Institute of Technology150001HarbinChina
| |
Collapse
|
20
|
Rübe CE, Raid S, Palm J, Rübe C. Radiation-Induced Brain Injury: Age Dependency of Neurocognitive Dysfunction Following Radiotherapy. Cancers (Basel) 2023; 15:cancers15112999. [PMID: 37296960 DOI: 10.3390/cancers15112999] [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] [Received: 04/20/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Cranial radiotherapy is a known risk factor for neurocognitive impairment in cancer survivors. Although radiation-induced cognitive dysfunction is observed in patients of all ages, children seem to be more vulnerable than adults to suffering age-related deficits in neurocognitive skills. So far, the underlying mechanisms by which IR negatively influences brain functions as well as the reasons for the profound age dependency are still insufficiently known. We performed a comprehensive Pubmed-based literature search to identify original research articles that reported on age dependency of neurocognitive dysfunction following cranial IR exposure. Numerous clinical trials in childhood cancer survivors indicate that the severity of radiation-induced cognitive dysfunction is clearly dependent on age at IR exposure. These clinical findings were related to the current state of experimental research providing important insights into the age dependency of radiation-induced brain injury and the development of neurocognitive impairment. Research in pre-clinical rodent models demonstrates age-dependent effects of IR exposure on hippocampal neurogenesis, radiation-induced neurovascular damage and neuroinflammation.
Collapse
Affiliation(s)
- Claudia E Rübe
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Silvia Raid
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Jan Palm
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| | - Christian Rübe
- Department of Radiation Oncology, Saarland University Medical Center, Kirrbergerstrasse Building 6.5, 66421 Homburg, Germany
| |
Collapse
|
21
|
Vielee ST, Wise JP. Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence. Brain Sci 2023; 13:500. [PMID: 36979310 PMCID: PMC10046019 DOI: 10.3390/brainsci13030500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Advancements in modern medicine have improved the quality of life across the globe and increased the average lifespan of our population by multiple decades. Current estimates predict by 2030, 12% of the global population will reach a geriatric age and live another 3-4 decades. This swelling geriatric population will place critical stress on healthcare infrastructures due to accompanying increases in age-related diseases and comorbidities. While much research focused on long-lived individuals seeks to answer questions regarding how to age healthier, there is a deficit in research investigating what aspects of our lives accelerate or exacerbate aging. In particular, heavy metals are recognized as a significant threat to human health with links to a plethora of age-related diseases, and have widespread human exposures from occupational, medical, or environmental settings. We believe heavy metals ought to be classified as a class of gerontogens (i.e., chemicals that accelerate biological aging in cells and tissues). Gerontogens may be best studied through their effects on the "Hallmarks of Aging", nine physiological hallmarks demonstrated to occur in aged cells, tissues, and bodies. Evidence suggests that cellular senescence-a permanent growth arrest in cells-is one of the most pertinent hallmarks of aging and is a useful indicator of aging in tissues. Here, we discuss the roles of heavy metals in brain aging. We briefly discuss brain aging in general, then expand upon observations for heavy metals contributing to age-related neurodegenerative disorders. We particularly emphasize the roles and observations of cellular senescence in neurodegenerative diseases. Finally, we discuss the observations for heavy metals inducing cellular senescence. The glaring lack of knowledge about gerontogens and gerontogenic mechanisms necessitates greater research in the field, especially in the context of the global aging crisis.
Collapse
Affiliation(s)
- Samuel T. Vielee
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - John P. Wise
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
22
|
Li X, Sheng J, Li Z, He Y, Zu Y, Li Y. Enhanced UV-B Radiation Induced the Proanthocyanidins Accumulation in Red Rice Grain of Traditional Rice Cultivars and Increased Antioxidant Capacity in Aging Mice. Int J Mol Sci 2023; 24:ijms24043397. [PMID: 36834809 PMCID: PMC9960751 DOI: 10.3390/ijms24043397] [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: 11/30/2022] [Revised: 01/10/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Proanthocyanidins are major UV-absorbing compounds. To clarify the effect of enhanced UV-B radiation on the proanthocyanidin synthesis and antioxidant capacity of traditional rice varieties in Yuanyang terraced fields, we studied the effects of enhanced UV-B radiation (0, 2.5, 5.0, 7.5 kJ·m-2·d-1) on the rice grain morphology, proanthocyanidins content, and synthesis. The effects of UV-B radiation on the antioxidant capacity of rice were evaluated by feeding aging model mice. The results showed that UV-B radiation significantly affected the grain morphology of red rice and increased the compactness of starch grains in the starch storage cells of central endosperm. The content of proanthocyanidin B2 and C1 in the grains was significantly increased by 2.5 and 5.0 kJ·m-2·d-1 UV-B radiation. The activity of leucoanthocyanidin reductase was higher in rice treated by 5.0 kJ·m-2·d-1 than other treatments. The number of neurons in the hippocampus CA1 of mice brain fed red rice increased. After 5.0 kJ·m-2·d-1 treatment, red rice has the best antioxidant effect on aging model mice. UV-B radiation induces the synthesis of rice proanthocyanidins B2 and C1, and the antioxidant capacity of rice is related to the content of proanthocyanidins.
Collapse
Affiliation(s)
- Xiang Li
- College of Resources and Environment, Yunnan Agricultural University, Kunming 650201, China
- National Engineering Research Center for Ornamental Horticulture, Flower Research Institute, Yunnan Academy of Agricultural Sciences, Kunming 650231, China
| | - Jianjun Sheng
- College of Resources and Environment, Yunnan Agricultural University, Kunming 650201, China
| | - Zuran Li
- College of Horticulture and Landscape, Yunnan Agricultural University, Kunming 650201, China
| | - Yongmei He
- College of Resources and Environment, Yunnan Agricultural University, Kunming 650201, China
| | - Yanqun Zu
- College of Resources and Environment, Yunnan Agricultural University, Kunming 650201, China
| | - Yuan Li
- College of Resources and Environment, Yunnan Agricultural University, Kunming 650201, China
- Correspondence:
| |
Collapse
|
23
|
Taylor BK, Smith OV, Miller GE. Chronic Home Radon Exposure Is Associated with Higher Inflammatory Biomarker Concentrations in Children and Adolescents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:246. [PMID: 36612568 PMCID: PMC9819293 DOI: 10.3390/ijerph20010246] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
Children are particularly vulnerable to the deleterious impacts of toxic environmental exposures, though the effects of some rather ubiquitous toxins have yet to be characterized in youths. One such toxin, radon gas, is known to accumulate to hazardous levels in homes, and has been linked with the incidence of lung cancer in aging adults. However, the degree to which chronic home radon exposure may impact risk for health problems earlier in life is unknown. Herein, we explored the degree to which chronic home radon exposure relates to biomarkers of low-grade inflammation in 68 youths ages 6- to 14 years old residing in an area of the United States prone to high home radon concentrations. Parents completed a home radon test kit, and youths provided a saliva sample to assess concentrations of five biomarkers. Using a multiple regression approach, we found that greater radon exposure was specifically associated with higher levels of C-reactive protein (β = 0.31, p = 0.007) and interleukin-1β (β = 0.33, p = 0.016). The data suggested specificity in associations between chronic home radon exposure and different biomarkers of inflammatory activity and highlight a pathway which may confer risk for future mental and physical health maladies.
Collapse
Affiliation(s)
- Brittany K. Taylor
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
- Department of Pharmacology and Neuroscience, Creighton University, Omaha, NE 68178, USA
| | - OgheneTejiri V. Smith
- Institute for Human Neuroscience, Boys Town National Research Hospital, Boys Town, NE 68010, USA
| | - Gregory E. Miller
- Institute for Policy Research and Department of Psychology, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
24
|
Chailapakul P, Kato TA. From Basic Radiobiology to Translational Radiotherapy. Int J Mol Sci 2022; 23:ijms232415902. [PMID: 36555542 PMCID: PMC9781393 DOI: 10.3390/ijms232415902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
The Special Issue, entitled "From basic radiobiology to translational radiotherapy", highlights recent advances in basic radiobiology and the potential to improve radiotherapy in translational research [...].
Collapse
|
25
|
Lee SH, Jeong YJ, Park J, Kim HY, Son Y, Kim KS, Lee HJ. Low-Dose Radiation Affects Cardiovascular Disease Risk in Human Aortic Endothelial Cells by Altering Gene Expression under Normal and Diabetic Conditions. Int J Mol Sci 2022; 23:8577. [PMID: 35955709 PMCID: PMC9369411 DOI: 10.3390/ijms23158577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
High doses of ionizing radiation can cause cardiovascular diseases (CVDs); however, the effects of <100 mGy radiation on CVD remain underreported. Endothelial cells (ECs) play major roles in cardiovascular health and disease, and their function is reduced by stimuli such as chronic disease, metabolic disorders, and smoking. However, whether exposure to low-dose radiation results in the disruption of similar molecular mechanisms in ECs under diabetic and non-diabetic states remains largely unknown; we aimed to address this gap in knowledge through the molecular and functional characterization of primary human aortic endothelial cells (HAECs) derived from patients with type 2 diabetes (T2D-HAECs) and normal HAECs in response to low-dose radiation. To address these limitations, we performed RNA sequencing on HAECs and T2D-HAECs following exposure to 100 mGy of ionizing radiation and examined the transcriptome changes associated with the low-dose radiation. Compared with that in the non-irradiation group, low-dose irradiation induced 243 differentially expressed genes (DEGs) (133 down-regulated and 110 up-regulated) in HAECs and 378 DEGs (195 down-regulated and 183 up-regulated) in T2D-HAECs. We also discovered a significant association between the DEGs and the interferon (IFN)-I signaling pathway, which is associated with CVD by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein−protein network analysis, and module analysis. Our findings demonstrate the potential impact of low-dose radiation on EC functions that are related to the risk of CVD.
Collapse
Affiliation(s)
- Soo-Ho Lee
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Ye Ji Jeong
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Jeongwoo Park
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Hyun-Yong Kim
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Yeonghoon Son
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Kwang Seok Kim
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Hae-June Lee
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| |
Collapse
|
26
|
Alessi I, Caroleo AM, de Palma L, Mastronuzzi A, Pro S, Colafati GS, Boni A, Della Vecchia N, Velardi M, Evangelisti M, Carboni A, Carai A, Vinti L, Valeriani M, Reale A, Parisi P, Raucci U. Short and Long-Term Toxicity in Pediatric Cancer Treatment: Central Nervous System Damage. Cancers (Basel) 2022; 14:cancers14061540. [PMID: 35326692 PMCID: PMC8946171 DOI: 10.3390/cancers14061540] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/11/2022] [Accepted: 03/14/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The purpose of this review is to describe central nervous system side effects in the treatment of pediatric cancer patients. Unfortunately, we must consider that the scarce data in the literature does not allow us to expand on some issues, especially those related to innovative immunotherapy. We have described the major neurotoxicities arising with the various types of treatment to help specialists who approach these treatments recognize them early, prevent them, and treat them promptly. Abstract Neurotoxicity caused by traditional chemotherapy and radiotherapy is well known and widely described. New therapies, such as biologic therapy and immunotherapy, are associated with better outcomes in pediatric patients but are also associated with central and peripheral nervous system side effects. Nevertheless, central nervous system (CNS) toxicity is a significant source of morbidity in the treatment of cancer patients. Some CNS complications appear during treatment while others present months or even years later. Radiation, traditional cytotoxic chemotherapy, and novel biologic and targeted therapies have all been recognized to cause CNS side effects; additionally, the risks of neurotoxicity can increase with combination therapy. Symptoms and complications can be varied such as edema, seizures, fatigue, psychiatric disorders, and venous thromboembolism, all of which can seriously influence the quality of life. Neurologic complications were seen in 33% of children with non-CNS solid malign tumors. The effects on the CNS are disabling and often permanent with limited treatments, thus it is important that clinicians recognize the effects of cancer therapy on the CNS. Knowledge of these conditions can help the practitioner be more vigilant for signs and symptoms of potential neurological complications during the management of pediatric cancers. As early detection and more effective anticancer therapies extend the survival of cancer patients, treatment-related CNS toxicity becomes increasingly vital. This review highlights major neurotoxicities due to pediatric cancer treatments and new therapeutic strategies; CNS primary tumors, the most frequent solid tumors in childhood, are excluded because of their intrinsic neurological morbidity.
Collapse
Affiliation(s)
- Iside Alessi
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Anna Maria Caroleo
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Luca de Palma
- Child Neurology Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Angela Mastronuzzi
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Stefano Pro
- Child Neurology Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | | | - Alessandra Boni
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Nicoletta Della Vecchia
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Margherita Velardi
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Melania Evangelisti
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Alessia Carboni
- Neuroradiology Unit, Imaging Department, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Andrea Carai
- Neurosurgery Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Luciana Vinti
- Department of Hematology/Oncology, Gene Therapy and Hematopoietic Transplantation, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Massimiliano Valeriani
- Child Neurology Unit, Department of Neuroscience, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Antonino Reale
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Pasquale Parisi
- Child Neurology, NESMOS Department, Faculty of Medicine and Psychology, Sant'Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| | - Umberto Raucci
- Department of Emergency, Acceptance and General Pediatrics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|