Ionizing radiation

电离辐射
  • 文章类型: Journal Article
    背景:三阴性乳腺癌(TNBC)是预后最差的乳腺癌亚型。放射治疗(RT)是疾病的核心方式之一;然而,RT的电离辐射具有严重的副作用。RT的一贯发展方向是以较低的辐射剂量达到更好的治疗效果。研究表明,将RT与非电离辐射疗法(如光和磁疗)结合使用可以实现协同作用,从而达到减少剂量和增强疗效的目的。
    方法:在本研究中,我们应用具有磁热功能的FeCoNP和光治疗剂IR-780来构建电离和非电离辐射协同纳米颗粒(INSNP)。INSNP首先进行形态学处理,尺寸,胶体稳定性,装载能力,和光热转换测试。随后,使用体外细胞系评估细胞抑制和细胞内化。在对NPs体内生物相容性进行全面评估后,建立荷瘤小鼠模型以评估其分布,有针对性的交付,和体内抗肿瘤作用。
    结果:INSNP的饱和磁化强度超过72emu/g,流体动力学粒径约为40nm,带负电荷的表面,良好的胶体稳定性和包封性能。INSNP在808nm处保持IR-780的光谱特性。在激光照射下,最高温度为92°C,INSNP还实现了体内有效的热温度。体内和体外试验均证明INSNP具有良好的生物相容性。INSNP在体内注射一次后仍有效超过一周,也可以通过永磁体引导和积聚在肿瘤中。稍后,结果表明,在低剂量RT和激光照射下,联合干预组表现出显著的协同作用,ROS产生率远高于RT组和光疗组。在小鼠模型中,60%的肿瘤被完全根除。
    结论:INSNPs有效地克服了RT治疗TNBC的许多缺点,为开发TNBC的新型临床治疗方法提供了实验依据。
    BACKGROUND: Triple-negative breast cancer (TNBC) is a subtype of breast cancer with the worst prognosis. Radiotherapy (RT) is one of the core modalities for the disease; however, the ionizing radiation of RT has severe side effects. The consistent development direction of RT is to achieve better therapeutic effect with lower radiation dose. Studies have demonstrated that synergistic effects can be achieved by combining RT with non-ionizing radiation therapies such as light and magnetic therapy, thereby achieving the goal of dose reduction and efficacy enhancement.
    METHODS: In this study, we applied FeCo NPs with magneto thermal function and phototherapeutic agent IR-780 to construct an ionizing and non-ionizing radiation synergistic nanoparticle (INS NPs). INS NPs are first subjected to morphology, size, colloidal stability, loading capacity, and photothermal conversion tests. Subsequently, the cell inhibitory and cellular internalization were evaluated using cell lines in vitro. Following comprehensive assessment of the NPs\' in vivo biocompatibility, tumor-bearing mouse model was established to evaluate their distribution, targeted delivery, and anti-tumor effects in vivo.
    RESULTS: INS NPs have a saturation magnetization exceeding 72 emu/g, a hydrodynamic particle size of approximately 40 nm, a negatively charged surface, and good colloidal stability and encapsulation properties. INS NPs maintain the spectral characteristics of IR-780 at 808 nm. Under laser irradiation, the maximum temperature was 92 °C, INS NPs also achieved the effective heat temperature in vivo. Both in vivo and in vitro tests have proven that INS NPs have good biocompatibility. INS NPs remained effective for more than a week after one injection in vivo, and can also be guided and accumulated in tumors through permanent magnets. Later, the results exhibited that under low-dose RT and laser irradiation, the combined intervention group showed significant synergetic effects, and the ROS production rate was much higher than that of the RT and phototherapy-treated groups. In the mice model, 60% of the tumors were completely eradicated.
    CONCLUSIONS: INS NPs effectively overcome many shortcomings of RT for TNBC and provide experimental basis for the development of novel clinical treatment methods for TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:环境低剂量辐射对放射从业人员的心血管系统影响仍不确定,需要进一步研究。本研究的目的是通过多组学联合综合基础实验,初步探讨低剂量辐射可能导致动脉粥样硬化的机制。
    方法:我们使用WGCNA和差异分析来鉴定辐射损伤和动脉粥样硬化测序数据集之间的共有基因和潜在通路,以及使用CIBERSORT反卷积算法进行组织转录组免疫浸润水平外推和单细胞转录组数据校正。通过将高脂饮食与5Gyγ射线全身低剂量电离辐射相结合来构建动物模型。通过酶联免疫吸附试验验证了NETs释放的检测。
    结果:分析揭示了辐射后和动脉粥样硬化两个数据集中的共有基因,提示免疫系统中性粒细胞可能是连接辐射和动脉粥样硬化的关键节点。NETs释放的中性粒细胞逝世亡能够影响动脉粥样硬化的发展。动物实验表明,低剂量辐射后中性粒细胞数量减少(P<0.05),NETs浓度降低,HF组NETs浓度显著升高(P<0.05)。与对照组相比,高脂饲料组血管内皮斑块明显增多,低剂量辐射组血管内皮斑块明显减少。
    结论:长期低剂量电离辐射照射刺激嗜中性粒细胞并抑制其产生NETs,导致动脉粥样硬化的抑制。
    OBJECTIVE: The cardiovascular system effects of environmental low-dose radiation exposure on radiation practitioners remain uncertain and require further investigation. The aim of this study was to initially investigate and explore the mechanisms by which low-dose radiation may contribute to atherosclerosis through a multi-omics joint comprehensive basic experiment.
    METHODS: We used WGCNA and differential analyses to identify shared genes and potential pathways between radiation injury and atherosclerosis sequencing datasets, as well as tissue transcriptome immune infiltration level extrapolation and single-cell transcriptome data correction using the CIBERSORT deconvolution algorithm. Animal models were constructed by combining a high-fat diet with 5 Gy γ-ray whole-body low-dose ionizing radiation. The detection of NETs release was validated by enzyme-linked immunosorbent assay.
    RESULTS: Analysis reveals shared genes in both datasets of post-irradiation and atherosclerosis, suggesting that immune system neutrophils may be a key node connecting radiation to atherosclerosis. NETs released by neutrophil death can influence the development of atherosclerosis. Animal experiments showed that the number of neutrophils decreased (P < 0.05) and the concentration of NETs reduced after low-dose radiation compared with the control group, and the concentration of NETs significantly increased (P < 0.05) in the HF group. Endothelial plaques were significantly increased in the high-fat feed group and significantly decreased in the low-dose radiation group compared with the control group.
    CONCLUSIONS: Long-term low-dose ionizing radiation exposure stimulates neutrophils and inhibits their production of NETs, resulting in inhibition of atherosclerosis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    电离辐射(IR)作为一种环境因素对线粒体构成了巨大的挑战,导致破坏细胞能量代谢并构成健康风险。尽管人们认识到IR对线粒体功能的有害影响,特定的分子靶标仍未完全阐明。在这项研究中,受γ射线作用的HeLa细胞表现出伴随的氧化应激,线粒体结构改变,ATP生产能力下降。γ射线诱导线粒体裂变的剂量依赖性诱导,同时表现为动态蛋白相关蛋白1(DRP1)的S616/S637磷酸化比率升高和线粒体融合蛋白mitofusin2(MFN2)的表达降低。DRP1敲除能有效减轻γ射线诱导的线粒体网络损伤,这意味着DRP1磷酸化可能是辐射诱导的线粒体损伤的效应子。线粒体外膜蛋白电压依赖性阴离子通道1(VDAC1)被确定为IR诱导的线粒体损伤的关键参与者。VDAC1抑制剂4,4'-二异硫氰基二苯乙烯-2,2'-二磺酸(DIDS),抵消了γ射线诱导的过度线粒体裂变,因此重新平衡糖酵解和氧化磷酸化平衡。发现这种代谢转变可以增强糖酵解能力,从而增强细胞弹性并提高癌细胞的放射敏感性。这些发现阐明了在辐射响应下控制线粒体形态的复杂调节机制。预期针对VDAC1的靶向药物的开发可能在增强肿瘤细胞对放射疗法和化学疗法的敏感性方面具有希望。
    The ionizing radiation (IR) represents a formidable challenge as an environmental factor to mitochondria, leading to disrupt cellular energy metabolism and posing health risks. Although the deleterious impacts of IR on mitochondrial function are recognized, the specific molecular targets remain incompletely elucidated. In this study, HeLa cells subjected to γ-rays exhibited concomitant oxidative stress, mitochondrial structural alterations, and diminished ATP production capacity. The γ-rays induced a dose-dependent induction of mitochondrial fission, simultaneously manifested by an elevated S616/S637 phosphorylation ratio of the dynamin-related protein 1 (DRP1) and a reduction in the expression of the mitochondrial fusion protein mitofusin 2 (MFN2). Knockdown of DRP1 effectively mitigated γ-rays-induced mitochondrial network damage, implying that DRP1 phosphorylation may act as an effector of radiation-induced mitochondrial damage. The mitochondrial outer membrane protein voltage-dependent anion channel 1 (VDAC1) was identified as a crucial player in IR-induced mitochondrial damage. The VDAC1 inhibitor 4,4\'-diisothiocyanatostilbene-2,2\'-disulfonic acid (DIDS), counteracts the excessive mitochondrial fission induced by γ-rays, consequently rebalancing the glycolytic and oxidative phosphorylation equilibrium. This metabolic shift was uncovered to enhance glycolytic capacity, thus fortifying cellular resilience and elevating the radiosensitivity of cancer cells. These findings elucidate the intricate regulatory mechanisms governing mitochondrial morphology under radiation response. It is anticipated that the development of targeted drugs directed against VDAC1 may hold promise in augmenting the sensitivity of tumor cells to radiotherapy and chemotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    目的:小肠是最容易受到电离辐射(IR)损伤的器官之一。然而,防止IR诱导的肠损伤的方法是有限的。CBLB502,一种来自沙门氏菌鞭毛蛋白的Toll样受体5(TLR5)激动剂,对各种组织和器官发挥辐射防护作用。然而,CBLB502对抗IR诱导的肠损伤的分子机制尚不清楚.因此,本研究旨在阐明IR诱导小鼠肠损伤的潜在机制以及CBLB502对这种情况的保护作用。
    方法:在不同时间点,在IR之前以不同剂量给予小鼠0.2mg/kgCBLB502,然后是存活率,体重,血象,并对小鼠进行组织病理学分析。
    结果:CBLB502减少IR诱导的肠损伤。RNA-seq分析显示不同剂量和持续时间的IR诱导不同的调控模式。CBLB502主要通过逆转IR诱导基因的表达并调节免疫过程和代谢途径来保护IR后的肠道损伤。
    结论:本研究初步阐述了CBLB502对IR诱导肠损伤的调控机制和潜在的分子保护机制,为鉴定介导IR诱导肠损伤保护的功能基因和分子机制提供了依据。
    OBJECTIVE: The small intestine is one of the organs most vulnerable to ionizing radiation (IR) damage. However, methods to protect against IR-induced intestinal injury are limited. CBLB502, a Toll-like receptor 5 (TLR5) agonist from Salmonella flagellin, exerts radioprotective effects on various tissues and organs. However, the molecular mechanisms by which CBLB502 protects against IR-induced intestinal injury remain unclear. Thus, this study aimed to elucidate the mechanisms underlying IR-induced intestinal injury and the protective effects of CBLB502 against this condition in mice.
    METHODS: Mice were administered 0.2 mg/kg CBLB502 before IR at different doses for different time points, and then the survival rate, body weight, hemogram, and histopathology of the mice were analyzed.
    RESULTS: CBLB502 reduced IR-induced intestinal injury. RNA-seq analysis revealed that different doses and durations of IR induced different regulatory patterns. CBLB502 protected against intestinal injury mainly after IR by reversing the expression of IR-induced genes and regulating immune processes and metabolic pathways.
    CONCLUSIONS: This study preliminarily describes the regulatory mechanism of IR-induced intestinal injury and the potential molecular protective mechanism of CBLB502, providing a basis for identifying the functional genes and molecular mechanisms that mediate protection against IR-induced injury.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    随着单细胞多组学的出现,结构细胞在免疫调节中的关键作用已经被揭示。但潜在的机制仍然知之甚少。这里,我们揭示了干扰素调节因子1(IRF1)的转录激活响应电离辐射,细胞毒性化学物质和SARS-CoV-2病毒感染决定了结构细胞的命运,并调节了结构细胞和免疫细胞之间的通讯。辐射诱导的mtDNA泄漏引发IRF1的核易位,使其能够调节炎症和细胞死亡相关基因的转录。鉴定了IRF1核定位序列(NLS)中的新翻译后修饰(PTM)位点。功能分析显示,NLS中乙酰化位点和磷酸化位点的突变阻断了IRF1的转录激活,并减少了响应电离辐射的细胞死亡。机械上,揭示了单链DNA传感器SSBP1和IRF1之间的相互调节,该调节抑制了辐射诱导的和STING/p300介导的IRF1PTM。此外,遗传缺失或药理学抑制IRF1温和辐射诱导的炎性细胞死亡,和辐射缓解剂还抑制SARS-CoV-2NSP-10介导的IRF1激活。因此,我们揭示了一种促进炎症的结构细胞中IRF1激活的新的细胞质导向机制,并强调了IRF1抑制剂对免疫疾病的潜在有效性.
    The key role of structural cells in immune modulation has been revealed with the advent of single-cell multiomics, but the underlying mechanism remains poorly understood. Here, we revealed that the transcriptional activation of interferon regulatory factor 1 (IRF1) in response to ionizing radiation, cytotoxic chemicals and SARS-CoV-2 viral infection determines the fate of structural cells and regulates communication between structural and immune cells. Radiation-induced leakage of mtDNA initiates the nuclear translocation of IRF1, enabling it to regulate the transcription of inflammation- and cell death-related genes. Novel posttranslational modification (PTM) sites in the nuclear localization sequence (NLS) of IRF1 were identified. Functional analysis revealed that mutation of the acetylation site and the phosphorylation sites in the NLS blocked the transcriptional activation of IRF1 and reduced cell death in response to ionizing radiation. Mechanistically, reciprocal regulation between the single-stranded DNA sensors SSBP1 and IRF1, which restrains radiation-induced and STING/p300-mediated PTMs of IRF1, was revealed. In addition, genetic deletion or pharmacological inhibition of IRF1 tempered radiation-induced inflammatory cell death, and radiation mitigators also suppressed SARS-CoV-2 NSP-10-mediated activation of IRF1. Thus, we revealed a novel cytoplasm-oriented mechanism of IRF1 activation in structural cells that promotes inflammation and highlighted the potential effectiveness of IRF1 inhibitors against immune disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在具有放射性物质和高能回旋加速器束的实验室中,对电离辐射的防护非常重要。位于宫城县东北大学的回旋加速器和放射性同位素中心(CYRIC),日本是著名的核科学实验室,拥有回旋加速器束和大量高活性放射性物质。考虑到这一点,重要的是要进行完整的辐射传输计算,以确保非职业和职业工人的安全。在目前的工作中,我们已经建立了一个完整的三维模型的主回旋加速器建筑和辐射实验室使用蒙特卡罗方法。我们发现,CYRIC建筑物内部和周围的分散光子和中子对职业和非职业工人没有重大风险。目前的工作和开发的模型将在辐射防护领域有用。
    Protection against ionizing radiations is important in laboratories with radioactive materials and high energy cyclotron beams. The Cyclotron and Radioisotope Center (CYRIC) located in Tohoku University in Miyagi prefecture, Japan and is a well-known nuclear science laboratory with cyclotron beams and substantial number of high activity radioactive materials. Considering this, it is important to perform complete radiation transport computations to ensure the safety of non-occupational and occupational workers. In the present work, we have developed a complete 3-dimensional model of the main cyclotron building and radiation labs using Monte Carlo method. We have found that the dispersed photons and neutrons inside and in the surrounding of the CYRIC building pose no significant risk to occupational and non-occupational workers. The present work and the developed models would be useful in the field of radiation protection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    聚乙二醇化超氧化物歧化酶(PEG-SOD)通常用作放射疗法中的细胞保护剂。然而,由于其皮肤渗透性差,其预防放射性皮炎的有效性有限。为了解决这个问题,开发了一种载有PEG-SOD的溶解微针(PSMN)贴片以有效预防放射性皮炎。最初,使用模板模具方法以聚乙烯吡咯烷酮K90作为基质材料制造PSMN贴片。PSMN表现出圆锥形状,具有足够的机械强度以穿透角质层。超过90%的PEG-SOD在30分钟内从PSMN贴剂中释放。值得注意的是,PSMN贴剂显示出明显高于PEG-SOD溶液的药物皮肤渗透,增加了500倍。在电脑模拟和皮肤药代动力学实验证实,PSMN贴剂增强药物渗透和皮肤吸收,与PEG-SOD溶液相反。更重要的是,PSMN贴片有效缓解电离辐射引起的皮肤损伤,加速受辐射影响的皮肤组织的愈合过程,并对皮肤组织中的DNA表现出高度有效的辐射防护活性。因此,PSMN贴剂是预防放射性皮炎的有希望的局部治疗方法。
    PEGylated superoxide dismutase (PEG-SOD) is commonly used as a cytoprotective agent in radiotherapy. However, its effectiveness in preventing radiation dermatitis is limited owing to its poor skin permeability. To address this issue, a PEG-SOD-loaded dissolving microneedle (PSMN) patch was developed to effectively prevent radiation dermatitis. Initially, PSMN patches were fabricated using a template mold method with polyvinylpyrrolidone K90 as the matrix material. PSMNs exhibited a conical shape with adequate mechanical strength to penetrate the stratum corneum. More than 90 % of PEG-SOD was released from the PSMN patches within 30 min. Notably, the PSMN patches showed a significantly higher drug skin permeation than the PEG-SOD solutions, with a 500-fold increase. In silico simulations and experiments on skin pharmacokinetics confirmed that PSMN patches enhanced drug permeation and skin absorption, in contrast to PEG-SOD solutions. More importantly, PSMN patches efficiently mitigated ionizing radiation-induced skin damage, accelerated the healing process of radiation-affected skin tissues, and exhibited highly effective radioprotective activity for DNA in the skin tissue. Therefore, PSMN patches are promising topical remedy for the prevention of radiation dermatitis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    电离辐射(IR)对自然环境和生物健康都构成重大威胁。在生物体发育早期暴露于特定剂量的电离辐射会导致发育毒性,特别是神经毒性。通过对非洲爪狼的实验(X.laevis),我们研究了辐射对早期发育阶段的影响。我们的发现表明,辐射以剂量依赖的方式导致X.laevis胚胎的发育异常和死亡,破坏氧化还原稳态并诱导细胞凋亡。此外,辐射引起神经毒性作用,导致胚胎中的异常行为和神经元损伤。对辐射诱导的神经毒性的潜在机制的进一步研究表明神经活性配体-受体相互作用途径的潜在参与,这得到了RNA-Seq分析的支持。与该途径相关的基因表达的验证和神经递质水平的分析证实了我们的假设。此外,我们通过依达拉奉抢救实验进一步验证了该信号通路在辐射诱导的神经毒性中的重要作用.本研究为辐射损伤研究建立了一个有价值的模型,并为辐射诱导的神经毒性机制提供了一些见解。
    Ionizing radiation (IR) poses a significant threat to both the natural environment and biological health. Exposure to specific doses of ionizing radiation early in an organism\'s development can lead to developmental toxicity, particularly neurotoxicity. Through experimentation with Xenopus laevis (X. laevis), we examined the effects of radiation on early developmental stage. Our findings revealed that radiation led to developmental abnormalities and mortality in X. laevis embryos in a dose-dependent manner, disrupting redox homeostasis and inducing cell apoptosis. Additionally, radiation caused neurotoxic effects, resulting in abnormal behavior and neuron damage in the embryos. Further investigation into the underlying mechanisms of radiation-induced neurotoxicity indicated the potential involvement of the neuroactive ligand-receptor interaction pathway, which was supported by RNA-Seq analysis. Validation of gene expression associated with this pathway and analysis of neurotransmitter levels confirmed our hypothesis. In addition, we further validated the important role of this signaling pathway in radiation-induced neurotoxicity through edaravone rescue experiments. This research establishes a valuable model for radiation damage studying and provides some insight into radiation-induced neurotoxicity mechanisms.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    不必要的电离辐射(IR)暴露通常会对正常细胞和器官造成急性和慢性氧化损伤。导致严重的生理甚至危及生命的后果。氨磷汀(AMF)是一种经过验证的辐射防护剂,广泛应用于放疗和化疗医学。但是短的半衰期限制了它的生物利用度和临床应用,仍然是一个巨大的挑战。DNA组装的纳米结构,特别是四面体框架核酸(tFNA)是具有卓越生物安全性的有前途的纳米载体,低生物毒性,运输效率高。tFNA还具有相对长期的维持结构稳定性和优异的内吞能力。因此,我们合成了一种基于tFNA的AMF递送系统,用于多器官辐射防护(tFNA@AMF,也称为纳米套装)。通过建立小鼠意外全身照射(TBI)模型和Lewis肺癌放疗模型,我们证明纳米套装可以通过调节抗凋亡和抗氧化应激的分子生物标志物来保护正常细胞免受IR诱导的DNA损伤。在意外全身照射(TBI)小鼠模型中,纳米套装预处理小鼠表现出令人满意的超氧化物歧化酶(SOD)活性和丙二醛(MDA)含量变化,和造血系统的功能恢复,减少IR引起的多器官病理损伤,保护小鼠免受致命辐射。更重要的是,在Lewis肺癌的放疗模型中,纳米套装显示了对正常器官的选择性放射防护,而没有肿瘤控制的干扰。基于一种方便的基于DNA四面体的纳米载体,这项工作提出了一种高效的AMF传递系统,具有延长的半衰期和增强的多器官辐射防护。这种纳米套装是一种有前途的策略,具有巨大的放射性保护临床翻译潜力。
    Unnecessary exposure to ionizing radiation (IR) often causes acute and chronic oxidative damages to normal cells and organs, leading to serious physiological and even life-threatening consequences. Amifostine (AMF) is a validated radioprotectant extensively applied in radiation and chemotherapy medicine, but the short half-life limits its bioavailability and clinical applications, remaining as a great challenge to be addressed. DNA-assembled nanostructures especially the tetrahedral framework nucleic acids (tFNAs) are promising nanocarriers with preeminent biosafety, low biotoxicity, and high transport efficiency. The tFNAs also have a relative long-term maintenance for structural stability and excellent endocytosis capacity. We therefore synthesized a tFNA-based delivery system of AMF for multi-organ radioprotection (tFNAs@AMF, also termed nanosuit). By establishing the mice models of accidental total body irradiation (TBI) and radiotherapy model of Lewis lung cancer, we demonstrated that the nanosuit could shield normal cells from IR-induced DNA damage by regulating the molecular biomarkers of anti-apoptosis and anti-oxidative stress. In the accidental total body irradiation (TBI) mice model, the nanosuit pretreated mice exhibited satisfactory alteration of superoxide dismutase (SOD) activities and malondialdehyde (MDA) contents, and functional recovery of hematopoietic system, reducing IR-induced pathological damages of multi-organ and safeguarding mice from lethal radiation. More importantly, the nanosuit showed a selective radioprotection of the normal organs without interferences of tumor control in the radiotherapy model of Lewis lung cancer. Based on a conveniently available DNA tetrahedron-based nanocarrier, this work presents a high-efficiency delivery system of AMF with the prolonged half-life and enhanced radioprotection for multi-organs. Such nanosuit pioneers a promising strategy with great clinical translation potential for radioactivity protection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:造血干细胞(HSC)再生是骨髓抑制造血恢复的基础,这是细胞毒性的威胁生命的副作用。骨髓抑制损伤后,HSC利基被严重破坏,虽然人们对生态位是否以及如何重塑和调节HSC再生知之甚少。
    方法:通过将小鼠暴露于亚致死剂量的电离辐射,建立了辐射损伤诱导的骨髓抑制的小鼠模型。数量的动态变化,通过流式细胞术确定HSCs和巨核细胞的分布和功能,免疫荧光,集落测定和骨髓移植,结合转录组学分析。使用共培养系统和过继转移确定HSC和巨核细胞之间的通讯。在体内和体外研究了信号机制,并使用巨核细胞特异性敲除小鼠和转基因小鼠进行巩固。
    结果:放射损伤后,巨核细胞成为HSC生态位的主要成分,并定位在更接近HSC的位置。同时,短暂性胰岛素样生长因子1(IGF1)的高分泌主要在辐射损伤后的巨核细胞中引起,而造血干细胞再生平行巨核细胞IGF1高分泌。机械上,HSC对巨核细胞IGF1分泌过多特别敏感,和IGF1信号下游的mTOR不仅促进HSC的激活,包括增殖和线粒体氧化代谢,但也抑制铁素吞噬限制HSC铁凋亡。因此,扩散之间的微妙协调,线粒体氧化代谢和铁凋亡确保了辐射损伤后功能性HSC的扩增。重要的是,在辐射损伤后,及时给予IGF1同时促进HSC再生和造血恢复,代表骨髓抑制的一种优越的治疗方法。
    结论:我们的研究确定巨核细胞是骨髓抑制性损伤的最后一道防线,巨核细胞IGF1是保护HSC再生的新生态位信号。
    BACKGROUND: Hematopoietic stem cell (HSC) regeneration underlies hematopoietic recovery from myelosuppression, which is a life-threatening side effect of cytotoxicity. HSC niche is profoundly disrupted after myelosuppressive injury, while if and how the niche is reshaped and regulates HSC regeneration are poorly understood.
    METHODS: A mouse model of radiation injury-induced myelosuppression was built by exposing mice to a sublethal dose of ionizing radiation. The dynamic changes in the number, distribution and functionality of HSCs and megakaryocytes were determined by flow cytometry, immunofluorescence, colony assay and bone marrow transplantation, in combination with transcriptomic analysis. The communication between HSCs and megakaryocytes was determined using a coculture system and adoptive transfer. The signaling mechanism was investigated both in vivo and in vitro, and was consolidated using megakaryocyte-specific knockout mice and transgenic mice.
    RESULTS: Megakaryocytes become a predominant component of HSC niche and localize closer to HSCs after radiation injury. Meanwhile, transient insulin-like growth factor 1 (IGF1) hypersecretion is predominantly provoked in megakaryocytes after radiation injury, whereas HSCs regenerate paralleling megakaryocytic IGF1 hypersecretion. Mechanistically, HSCs are particularly susceptible to megakaryocytic IGF1 hypersecretion, and mTOR downstream of IGF1 signaling not only promotes activation including proliferation and mitochondrial oxidative metabolism of HSCs, but also inhibits ferritinophagy to restrict HSC ferroptosis. Consequently, the delicate coordination between proliferation, mitochondrial oxidative metabolism and ferroptosis ensures functional HSC expansion after radiation injury. Importantly, punctual IGF1 administration simultaneously promotes HSC regeneration and hematopoietic recovery after radiation injury, representing a superior therapeutic approach for myelosuppression.
    CONCLUSIONS: Our study identifies megakaryocytes as a last line of defense against myelosuppressive injury and megakaryocytic IGF1 as a novel niche signal safeguarding HSC regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号