Normal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (2024)

  • Journal List
  • Nan Fang Yi Ke Da Xue Xue Bao
  • v.44(5); 2024 May 20
  • PMC11166715

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Normal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (1)

Link to Publisher's site

Nan Fang Yi Ke Da Xue Xue Bao. 2024 May 20; 44(5): 801–809.

PMCID: PMC11166715

PMID: 38862437

Language: English | Chinese

YUAN Tong, GUO Yuying, ZHANG Junling,Normal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (2) and FAN SaijunNormal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (3)

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

Objective

To evaluate the therapeutic effect of normal mouse serum on radiation pneumonitis in mice and explore the possible mechanism.

Methods

Mouse models of radiation pneumonitis induced by thoracic radiation exposure were given intravenous injections of 100 μL normal mouse serum or normal saline immediately after the exposure followed by injections once every other day for a total of 8 injections. On the 15th day after irradiation, histopathological changes of the lungs of the mice were examined using HE staining, the levels of TNF-α, TGF-β, IL-1α and IL-6 in the lung tissue and serum were detected using ELISA, and the percentages of lymphocytes in the lung tissue were analyzed with flow cytometry. High-throughput sequencing of exosome miRNA was carried out to explore the changes in the signaling pathways. The mRNA expression levels of the immune-related genes were detected by qRT-PCR, and the protein expressions of talin-1, tensin2, FAK, vinculin, α-actinin and paxillin in the focal adhesion signaling pathway were detected with Western blotting.

Results

In the mouse models of radiation pneumonitis, injections of normal mouse serum significantly decreased the lung organ coefficient, lowered the levels of TNF-α, TGF-β, IL-1α and IL-6 in the serum and lung tissues, and ameliorated infiltration of CD45+, CD4+ and Treg lymphocytes in the lung tissue (all P<0.05). The expression levels of Egfr and Pik3cd genes at both the mRNA and protein levels and the protein expressions of talin-1, tensin2, FAK, vinculin, α‑actinin and paxillin were all significantly down-regulated in the mouse models after normal mouse serum treatment.

Conclusion

Normal mouse serum ameliorates radiation pneumonitis in mice by inhibiting the expressions of key proteins in the Focal adhesion signaling pathway.

Keywords: normal mouse serum, radiation pneumonitis, exosomes, focal adhesion pathway

Abstract

目的

探讨正常小鼠血清(NMS)对放射性肺炎的治疗作用及可能机制。

方法

建立胸腔照射诱导的放射性肺炎模型,将小鼠分为对照组、静脉注射血清组、照射组和照射后静脉注射血清组。注射血清组小鼠在照射后立即静脉注射正常小鼠血清100 μL,对照组小鼠注射100 μL 生理盐水,隔天注射1次,共注射8次。照射后15 d取材,HE染色检测肺组织形态学变化,ELISA检测小鼠肺组织和血清中炎症因子肿瘤坏死因子-α(TNF-α)、转化生长因子-β(TGF-β)、白细胞介素-1α(IL-1α)、白细胞介素-6(IL-6)水平;流式细胞术检测肺组织内淋巴细胞比例变化。外泌体miRNA高通量测序探索处理后小鼠的信号通路变化,qRT-PCR检测免疫相关基因的表达水平,使用Western blotting检测黏着斑通路talin-1、tensin 2、FAK、vinculin、α-actinin和paxillin蛋白的表达。

结果

与照射组相比,照射后注射血清组小鼠肺脏器系数、血清及肺组织上清液中炎症因子TNF-α、TGF-β、IL-1α、IL-6水平显著降低(P<0.05),CD45+、CD4+、Treg淋巴细胞在小鼠肺组织中的浸润程度显著下降(P<0.05);肺中EgfrPik3cd的mRNA和蛋白表达水平显著下调,talin-1、tensin 2、FAK、vinculin、α-actinin和paxillin蛋白的表达水平也显著降低(P<0.05)。

Keywords: 正常小鼠血清, 放射性肺炎, 外泌体, 粘着斑途径

Original Article

INTRODUCTION

Lung cancer is one of the leading causes of cancer-related deaths worldwide with a high mortality rate1. Thoracic radiotherapy is the primary treatment option for lung cancer, but its administration is associated with potential complications such as acute radiation pneumonitis, which seriously affects the patients' quality of life 2-4. Lung tissues are sensitive to ionizing radiation and vulnerable to radiation injuries that causes release of proinflammatory cytokines, which leads to recruitment of inflammatory cells to the alveoli and interstitium and hence results in acute radiation pneumonitis 56. It is estimated that up to 50% lung cancer patients develop radiation pneumonitis following thoracic radiotherapy7. The exact pathogenic mechanism of radiation pneumonitis remains to be fully elucidated, and effective clinical interventions are currently unavailable.

Our previous work demonstrated that normal mouse serum (NMS) could alleviate total body irradiation-induced hematopoietic system injury in mice by improving the systemic environment, in which process exosomes, a key active component in the serum, play an important protective role8. In addition to its anti-inflammatory effects910 and ability to penetrate the blood-brain barrier11, exosomes have been shown to participate in various biological processes including tumor cell migration1213, antigen presentation14, cell apoptosis15, immune responses16, and intercellular signaling17. In this study, we aimed to investigate the protective effect of intravenous injection of NMS against radiation-induced acute lung injury in mice and explore the mechanisms that mediate this effect by sequencing analysis of miRNAs in serum-derived exosomes, KEGG pathway enrichment analysis, and validation of the protein expressions.

METHODS

Animals and NMS collection

All the animal experiments were approved by the Ethics Committee for Laboratory Animals of the Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College (IRM/2-IACUC-2021222), and were conducted in line with the regulations regarding the welfare and ethics of laboratory animals in China. Male C57BL/6 mice (6-8 weeks old, body weight ~20 g) were obtained from Beijing HFK Biotechnology Co., Ltd. and housed under specific pathogen-free (SPF) conditions in the Animal Experimental Center of the Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College. Twenty-four mice with a similar genetic background were used for collecting orbital blood, which was refrigerated at 4 ℃ for 1-2 h followed by centrifugation at 4000 r/min for 10 min to isolate the serum.

Animal grouping and treatment

Twenty-four mice were randomly divided into control group, serum injection group, irradiation (IR) model group, and IR+serum injection group (n=6). In serum injection group and IR+serum injection group, the mice were subjected to intravenous injection of 100 μL of NMS via the tail vein immediately after irradiation exposure, and those in the control group and IR model group received injection of 100 μL physiological saline. The injections were administered every other day for a total of 8 times, and the mice were euthanized 15 days after the irradiation for examination.

Irradiation protocol

To simulate thoracic radiotherapy in lung cancer patients, the mice were anesthetized and the thorax of mice was exposed to the irradiation source, with the rest of the body shielded from radiation by a lead block. The irradiation was performed using the Gammacell®40 Exactor irradiation apparatus (Atomic Energy Canada Ltd.) with a total irradiation dose of 13 Gy delivered at a dose rate of 0.88 Gy/min. The mice in the control group were sham-irradiated.

Lung/body coefficient measurement

On the 15th day after irradiation, the mice were euthanized and the lungs were dissected and weighed using a precision electronic balance. The lung/body coefficients were calculated as follows: Lung weight/ Body weight × 100%.

Flow cytometry analysis

Fresh mouse lung tissue samples were placed in a sterile dish and cut into small pieces. The tissues were treated with a preheated enzyme mixture containing Liberase TM Research Grade (Roche), DNase I (Roche) and DMEM (gibco) at 37 ℃ for 30 min, and the single cells were collected. The cell concentration was adjusted to 5×106/mL, and 200 μL of the cell suspension was stained with PerCP/Cyanine5.5 anti-mouse CD45 antibody (BioLegend), CD4 monoclonal antibody (GK1.5), PE (eBioscience), APC anti-mouse CD25 antibody (BioLegend), and Pacific Blue™ anti-mouse FOXP3 antibody (BioLegend) for flow cytometry analysis using BD FACSCelesta flow cytometer (BD Bioscience, San Jose, CA, USA), and the results were analyzed using FlowJo software.

Enzyme linked immunosorbent assay (ELISA)

ELISA was used to measure the levels of inflammatory factors in both the lung tissue and serum of the mice. Lung tissue hom*ogenate was prepared using a tissue crusher (TissuePrep) and centrifuged to collect the supernatant. ELISA measurement was performed using an ELISA kit (Shanghai Enzyme-linked Biotechnology Co., Ltd.) following the manufacturer's instructions. Briefly, the diluted sample and biotin-labeled anti-TXA2 antibody were both added into the wells of the ELISA plate, and after incubation at 37 ℃ for 1 h, the plate was washed 3 times followed by addition of HRP-conjugated affinase. The mixture was gently shaken and incubated at 37 ℃ for 30 min, and following washing for 3 times, the substrates A and B were added. The plate was incubated at 37 ℃ for 10 min in darkness followed immediately by addition of the termination solution. The absorbance values at 450 nm of each well were measured using a micro-plate reader (Rayto, RT-6500).

HE staining

Mouse lung tissues were fixed in 4% paraformaldehyde overnight and embedded in paraffin. The paraffin sections (4 μm) were deparaffinized in xylene, rehydrated in a concentration gradient of alcohol, stained with hematoxylin, and dehydrated with gradient ethanol followed by xylene permeation and neutral resin sealing. The sections were then observed under an automatic digital slice scanning system (Beijing Uniona Technology Co., PRECICE 510).

Western blotting

Lung tissue proteins were extracted using tissue RIPA lysate with 1% protease inhibitor. Following polyacrylamide gel electrophoresis, the proteins were transferred onto a PVDF membrane, coated with the corresponding antibodies, and visualized using a Bio-Rad gel imaging system. The antibodies used in the experiment included anti-paxillin antibody (Abcam, 1∶1000), HRP-conjugated Affinipure goat anti-rabbit IgG (H+L) (Proteintech, 1∶5000), HRP-conjugated Affinipure goat anti-mouse IgG (H+L) (Proteintech, 1∶5000), Focal Adhesion Protein Antibody Sampler Kit (Cell Signaling Technology, 1∶1000), Anti-EGFR antibody (Abcam, 1∶2000), anti-PI3 kinase p110 delta antibody (Abcam, 1∶2000), and β-tubulin monoclonal antibody (Proteintech, 1∶20 000).

Extraction and sequencing analysis of exosomes

Exosomes were isolated from the blood of the mice from the 4 groups using an exosome extraction and purification Kit (Umibio Science and Technology Co., Ltd, Shanghai, China). Sequencing analysis was performed by Beijing Genomics Institution (BGI) and further data mining was carried out using the BGI Multi-Omics System.

Statistical analysis

Statistical analysis was performed using GraphPad Prism 8. The data were expressed as Mean±SD and analyzed using one-way ANOVA. Turkey's multiple comparison test was used to compare the data between two groups, and P<0.05 was considered to indicate a statistically significant difference.

RESULTS

NMS ameliorates radiation-induced structural damage of mouse lungs

The mice in the control group showed normal morphology of the bilateral lungs, which were soft with a smooth surface and good elasticity without congestion or edema (Fig.1A), and the thin alveolar walls were intact without inflammatory cell infiltration (Fig.1C). In contrast, the lungs of the irradiated mice were hard and dark red in color, showing obvious edema and congestion with increased volume and organ coefficient (P<0.001, Fig.1B). The lungs of irradiated mice showed severely damaged structure with interstitial edema, obvious inflammatory cell infiltration, thickening of the alveolar walls and reduced airspace volume. Intravenous injections of NMS significantly improved radiation-induced lung tissue damage and decreased the lung coefficient, and the lung tissue histology was almost normal without noticeable inflammatory cell infiltration.

Open in a separate window

Fig.1

Normal mouse serum effectively protects normal histological structure of the lung tissue of irradiated mice. A: Gross observation of mouse lungs. B: Comparison of lung organ coefficients among the 4 groups. C: HE staining of the lung tissues in the 4 groups. Data are presented as Mean±SD (n=5). *P<0.05, ***P<0.001.

NMS lowers inflammatory cytokine levels in irradiated mice

Fig.2 shows that compared with those in the control group, TGF-β, TNF-α, IL-1α, and IL-6 levels in both the lung tissues and serum were significantly elevated in IR model group, but NMS injections effectively lowered the levels of these cytokines in the irradiated mice.

Open in a separate window

Fig.2

Intravenous injections of normal mouse serum down-regulate the levels of inflammatory factors in the lung tissue and serum of irradiated mice. A-D: Levels of TGF-β, TNF-α, IL-1α, and IL-6 in the lung tissue, respectively. E-H: Serum levels of TGF-β, TNF-α, IL-1α, and IL-6 in the serum, respectively. Data are presented as Mean±SD (n=8 in panel A and panel E, and n=5 in the rest panels). *P<0.05, **P<0.01, ***P<0.001.

NMS reduces lymphocyte infiltration in the lungs of irradiated mice

Compared with the control mice, the irradiated mice showed significantly increased percentages of CD45+, CD4+ and Treg cells in the lung tissue on day 15 after irradiation. The percentages of CD45+, CD4+ and Treg cells were all significantly lowered in the mouse models receiving NMS injections (Fig.3).

Open in a separate window

Fig.3

NMS reduces lymphocyte infiltration in the lung tissue of irradiated mice A-D: Results of fluorescence-activated cell sorting showing gating for CD45+, CD4+ and Treg cells and their percentages in total cells. Data are presented as Mean±SD (n=5). *P<0.05, **P<0.01.

Differential miRNAs in irradiated and NMS-treated mice

The results of RNAseq showed that compared with the control group, 161 miRNA were up-regulated and 76 miRNA were down-regulated significantly in irradiated mice; compared with the IR group, 49 miRNA were up-regulated and 170 were down-regulated in IR+serum group (Fig.4A). The Venn diagram shows 25 common differential miRNA among the 3 groups (Fig.4B).Immune-related target genes of the differential miRNAs

Open in a separate window

Fig.4

Statistics of the differential miRNAs in irradiated and NMS-treated mice (A) and Venn diagram of the differential miRNAs (B).

Data mining using the BGI Multi-Omics System under the condition "log2[(IR+Serum)/IR]<2, log2(IR/control) >2" identified 16 miRNAs among the 25 common differential miRNAs (Tab.1). These 16 miRNAs involved a total of 615 target genes, among which KEGG pathway enrichment analysis identified 6 immune-related differential genes (Gsk3b, Prkci, Cul3, Tiam1, Egfr, and Pik3cd; Fig.5A). Verification with qRT-PCR showed that Egfr and Pik3cd mRNA expressions were significantly increased in the lung tissue of irradiated mice, and their expressions were obviously lowered by NMS injections (Fig.5B, C). Western blotting also confirmed significant upregulation of EGFR and P110δ (the protein encoded by Pik3cd) expressions in irradiated mice and their reduction following NMS treatment (Fig.5D, E).

Tab.1

miRNAs with significant differential expressions in irradiated and NMS-treated mice

Normal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (8)

Open in a separate window

Open in a separate window

Fig.5

Number of immune-related differential genes and their expression at mRNA and protein levels in irradiated and NMS-treated mice. A: Classification of KEGG pathways. B, C: Relative mRNA expressions of Gsk3b, Cul3, Tiam1, Prkci, Egfr and Pik3cd in the lung tissue. D, E: Western blots of EGFR and P110δ proteins and their relative expression levels. Data are presented as Mean±SD (n=5 in panel B and panel C, and n=3 in panel D and panel E). *P<0.05, **P<0.01, ***P<0.001.

NMS down-regulates focal adhesion pathway in irradiated mice

Fig.6A shows the results of KEGG pathway enrichment analysis of the 615 target genes. The focal adhesion pathway, which is closely related to inflammation and fibrosis, enriched the greatest number of the target genes. Western blot analysis of the proteins related to this pathway showed that the expression levels of talin-1, tensin 2, FAK, vinculin, α-actinin and paxillin (P<0.05 or 0.01) were all significantly increased in the lung tissue of the irradiated mice as compared with the control mice. These proteins were all significantly down-regulated in irradiated mice with NMS injections (P<0.05; Fig.6B-D).

Open in a separate window

Fig.6

Normal mouse serum significantly down-regulates the expression of Focal adhesion pathway-related proteins in irradiated mice. A: Enrichment bubble chart of KEGG pathway. B-D: Western blotting of the key proteins in the focal adhesion signaling pathway and their relative expression levels in the lung tissues. Data are presented as Mean±SD (n=3). *P<0.05, **P<0.01, ***P<0.001.

DISCUSSION

Here we demonstrated that NMS offers protection against radiation-induced acute lung injuries in mice by preserving normal histology and ameliorating inflammation in the lung tissues. Pulmonary inflammation is a severe complication following thoracic radiotherapy as a result of ionizing radiation-induced DNA damage and intracellular reactive oxygen species accumulation in the lung cells18-21, which release cytokines to attract the inflammatory cells to the alveoli and interstitium622. We found that intravenous injections of NMS could effectively reduce the levels of the inflammatory cytokines (TNF-α, TGF-β, IL-1α, and IL-6) in lung tissues and serum of the irradiated mice, suggesting that NMS can significantly reduce inflammatory responses to radiation exposure and further, its crucial role in maintaining homeostasis within the lung tissue of irradiated mice.

We examined the effect of NMS treatment on CD45+, CD4+T, and Treg cell infiltration in the lung tissues of the irradiated mice. CD45 is marker of infiltrating leukocytes, CD4+T cells represent a comprehensive group of T helper cells, and Treg cells, as a subset of CD4+T cells, perform immune-suppressive functions and play a vital role in preventing excessive immune responses. Our results demonstrated that NMS significantly diminished lymphocyte infiltration in the lung tissues of irradiated mice. Radiation exposure caused a marked increase of Treg cells in mouse lung tissues, which was an auto-immune regulatory response to prevent excessive immune responses to the exposure. NMS treatment alleviated radiation pneumonitis and also significantly lowered the percentage of Treg cells in the lung tissue of irradiated mice. These findings provide further evidence that NMS alleviates inflammatory symptoms in mice with radiation pneumonitis and maintains the immune balance in radiation-damaged lung tissues.

We further explored the mechanism underlying the protective effects of NMS against radiation pneumonitis in mice. In a previous work we investigated the changes in the serum proteome in mice with 4 Gy whole-body radiation, and found that treatment of the irradiated mice with NMS resulted in changes in the proteins associated with acute stress (such as SAA2, APCS, and immunoglobulins)8, but given the complex composition of serum, we were unable to determine the mechanisms by which NMS induced these changes. Exosomes are crucial mediators for executing biological functions of the serum and play significant roles in tumor cell metastasis1223, antigen presentation24, inhibition of inflammation and fibrosis25-29, and intercellular signaling1730. Signal transduction of the exosome mainly relies on the encapsulated miRNAs, which regulate their translation of their target genes by modulating mRNA stability. Our previous work have demonstrated the significant role in mediating the effects of NMS for alleviating ionizing radiation-induced injury of the hematopoietic system8. Li et al31 also reported that serum-derived exosomes could effectively alleviate radiation-induced intestinal damage.

Based on these observations, we explored the mechanism of NMS against radiation pneumonitis through sequencing analysis of the miRNAs in the exosomes. The results revealed 25 common differentially expressed miRNAs in irradiated and NMS-treated mice and two immune- and inflammation-related differentially expressed genes, namely Egfr and Pik3cd. The focal adhesion signaling pathway was identified as the likely mechanism that mediates the protective effects of NMS against radiation pneumonitis in mice. Previous studies have shown that the key enzymes in this pathway play essential roles in relieving inflammation, mitigating fibrosis, and regulating immunity3233. We show in this study that intravenous injection of NMS following radiation exposure significantly lowers the expressions of the key proteins in the focal adhesion signaling pathway including talin-1, tensin 2, FAK, vinculin, α-actinin, and paxillin, which supports the notion that the focal adhesion signaling pathway plays a key role in mediating the therapeutic effect of NMS for radiation pneumonitis in mice.

In conclusion, we demonstrate in this study that NMS can effectively alleviate radiation pneumonitis in mice early after radiation exposure, and the therapeutic effect is possibly achieved by inhibiting the focal adhesion signaling pathway in the lung tissues. These findings provide a new perspective for clinical treatment of radiation pneumonitis.

Funding Statement

中国医学科学院医学与健康科技创新工程重大协同创新项目(2021-I2M-1-042);中国医学科学院国家医学健康科技战略平台与体系建设项目(2022-I2M-2-003);国家自然科学基金(82273577)

Supported by the CAMS Innovation Fund for Medical Sciences (CIFMS, 2021-I2M-1-042, 2022-I2M-2-003) and National Natural Science Foundation of China (82273577).

REFERENCES

1. Sung H, Ferlay J, Siegel RL, et al.. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2021, 71(3): 209-49. DOI: 10.3322/caac.21660 [PubMed] [CrossRef] [Google Scholar]

2. Arora A, Bhuria V, Singh S, et al.. Amifostine analog, DRDE-30, alleviates radiation induced lung damage by attenuating inflammation and fibrosis[J]. Life Sci, 2022, 298: 120518. DOI: 10.1016/j.lfs.2022.120518 [PubMed] [CrossRef] [Google Scholar]

3. Hanania AN, Mainwaring W, Ghebre YT, et al.. Radiation-induced lung injury: assessment and management[J]. Chest, 2019, 156(1): 150-62. DOI: 10.1016/j.chest.2019.03.033 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

4. Giridhar P, Mallick S, Rath GK, et al.. Radiation induced lung injury: prediction, assessment and management[J]. Asian Pac J Cancer Prev, 2015, 16(7): 2613-7. DOI: 10.7314/apjcp.2015.16.7.2613 [PubMed] [CrossRef] [Google Scholar]

5. Citrin DE, Shankavaram U, Horton JA, et al.. Role of type II pneumocyte senescence in radiation-induced lung fibrosis[J]. J Natl Cancer Inst, 2013, 105(19): 1474-84. DOI: 10.1093/jnci/djt212 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

6. Abratt RP, Morgan GW, Silvestri G, et al.. Pulmonary complications of radiation therapy[J]. Clin Chest Med, 2004, 25(1): 167-77. DOI: 10.1016/s0272-5231(03)00126-6 [PubMed] [CrossRef] [Google Scholar]

7. Gao J, Peng S, Shan XN, et al.. Inhibition of AIM2 inflammasome-mediated pyroptosis by Andrographolide contributes to amelioration of radiation-induced lung inflammation and fibrosis[J]. Cell Death Dis, 2019, 10(12): 957. DOI: 10.1038/s41419-019-2195-8 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

8. Zhang JL, Han XD, Zhao Y, et al.. Mouse serum protects against total body irradiation-induced hematopoietic system injury by improving the systemic environment after radiation[J]. Free Radic Biol Med, 2019, 131: 382-92. DOI: 10.1016/j.freeradbiomed.2018.12.021 [PubMed] [CrossRef] [Google Scholar]

9. He CJ, Zheng S, Luo Y, et al.. Exosome theranostics: biology and translational medicine[J]. Theranostics, 2018, 8(1): 237-55. DOI: 10.7150/thno.21945 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

10. Kalluri R, LeBleu VS. The biology, function, and biomedical applications of exosomes[J]. Science, 2020, 367(6478): eaau6977. DOI: 10.1126/science.aau6977 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

11. Gurunathan S, Kang MH, Jeyaraj M, et al.. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes[J]. Cells, 2019, 8(4): 307. DOI: 10.3390/cells8040307 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

12. Hood JL, San RS, Wickline SA. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis[J]. Cancer Res, 2011, 71(11): 3792-801. DOI: 10.1158/0008-5472.can-10-4455 [PubMed] [CrossRef] [Google Scholar]

13. Zhang L, Yu DH. Exosomes in cancer development, metastasis, and immunity[J]. Biochim Biophys Acta Rev Cancer, 2019, 1871(2): 455-68. DOI: 10.1016/j.bbcan.2019.04.004 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

14. Bobrie A, Colombo M, Raposo G, et al.. Exosome secretion: molecular mechanisms and roles in immune responses[J]. Traffic, 2011, 12(12): 1659-68. DOI: 10.1111/j.1600-0854.2011.01225.x [PubMed] [CrossRef] [Google Scholar]

15. Gregory CD, Pound JD. Microenvironmental influences of apoptosis in vivo and in vitro[J]. Apoptosis, 2010, 15(9): 1029-49. DOI: 10.1007/s10495-010-0485-9 [PubMed] [CrossRef] [Google Scholar]

16. Howitt J, Hill AF. Exosomes in the pathology of neurodegenerative diseases[J]. J Biol Chem, 2016, 291(52): 26589-97. DOI: 10.1074/jbc.r116.757955 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

17. Harding CV, Heuser JE, Stahl PD. Exosomes: looking back three decades and into the future[J]. J Cell Biol, 2013, 200(4): 367-71. DOI: 10.1083/jcb.201212113 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

18. Kang H, Kim SC, Oh Y. Fucoxanthin abrogates ionizing radiation-induced inflammatory responses by modulating sirtuin 1 in macrophages[J]. Mar Drugs, 2023, 21(12): 635. DOI: 10.3390/md21120635 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

19. Téoule R. Radiation-induced DNA damage and its repair[J]. Int J Radiat Biol Relat Stud Phys Chem Med, 1987, 51(4): 573-89. DOI: 10.1080/09553008414552111 [PubMed] [CrossRef] [Google Scholar]

20. Zheng Y, Li H, Bo XC, et al.. Ionizing radiation damage and repair from 3D-genomic perspective[J]. Trends Genet, 2023, 39(1): 1-4. DOI: 10.1016/j.tig.2022.07.004 [PubMed] [CrossRef] [Google Scholar]

21. Zhang JX, Wang XL, Vikash V, et al.. ROS and ROS-mediated cellular signaling[J]. Oxid Med Cell Longev, 2016, 2016: 4350965. DOI: 10.1155/2016/4350965 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

22. Xu TK, Zhang YY, Chang PY, et al.. Mesenchymal stem cell-based therapy for radiation-induced lung injury[J]. Stem Cell Res Ther, 2018, 9(1): 18. DOI: 10.1186/s13287-018-0776-6 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

23. Milane L, Singh A, Mattheolabakis G, et al.. Exosome mediated communication within the tumor microenvironment[J]. J Control Release, 2015, 219: 278-94. DOI: 10.1016/j.jconrel.2015.06.029 [PubMed] [CrossRef] [Google Scholar]

24. McCall CE, El Gazzar M, Liu TF, et al.. Epigenetics, bioenergetics, and microRNA coordinate gene-specific reprogramming during acute systemic inflammation[J]. J Leukoc Biol, 2011, 90(3): 439-46. DOI: 10.1189/jlb.0211075 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

25. Wang CL, Xu MC, Fan QZ, et al.. Therapeutic potential of exosome-based personalized delivery platform in chronic inflammatory diseases[J]. Asian J Pharm Sci, 2023, 18(1): 100772. DOI: 10.1016/j.ajps.2022.100772 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

26. Zheng X, Zhao D, Liu Y, et al.. Regeneration and anti-inflammatory effects of stem cells and their extracellular vesicles in gynecological diseases[J]. Biomed Pharmacother, 2023, 168: 115739. DOI: 10.1016/j.biopha.2023.115739 [PubMed] [CrossRef] [Google Scholar]

27. Liu C, Xiao K, Xie LX. Advances in the use of exosomes for the treatment of ALI/ARDS[J]. Front Immunol, 2022, 13: 971189. DOI: 10.3389/fimmu.2022.971189 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

28. Xu CJ, Zhao J, Li QY, et al.. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model[J]. Stem Cell Res Ther, 2020, 11(1): 503. DOI: 10.1186/s13287-020-02023-9 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

29. Noonin C, Thongboonkerd V. Exosome-inflammasome crosstalk and their roles in inflammatory responses[J]. Theranostics, 2021, 11(9): 4436-51. DOI: 10.7150/thno.54004 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

30. Sun ZQ, Yang SX, Zhou QB, et al.. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment[J]. Mol Cancer, 2018, 17(1): 82. DOI: 10.1186/s12943-018-0831-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]

31. Li H, Zhao SY, Jiang M, et al.. Biomodified extracellular vesicles remodel the intestinal microenvironment to overcome radiation enteritis[J]. ACS Nano, 2023, 17(14): 14079-98. DOI: 10.1021/acsnano.3c04578 [PubMed] [CrossRef] [Google Scholar]

32. Wong VW, Rustad KC, Akaishi S, et al.. Focal adhesion kinase links mechanical force to skin fibrosis via inflammatory signaling[J]. Nat Med, 2011, 18(1): 148-52. DOI: 10.1038/nm.2574 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

33. Chen WC, Yu WK, Su VYF, et al.. NLRP3 inflammasome activates endothelial-to-mesenchymal transition via focal adhesion kinase pathway in bleomycin-induced pulmonary fibrosis[J]. Int J Mol Sci, 2023, 24(21): 15813. DOI: 10.3390/ijms242115813 [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from Journal of Southern Medical University are provided here courtesy of Editorial Department of Journal of Southern Medical University

Normal mouse serum alleviates radiation pneumonitis in mice by inhibiting the focal adhesion signaling pathway (2024)
Top Articles
Latest Posts
Article information

Author: Ray Christiansen

Last Updated:

Views: 6284

Rating: 4.9 / 5 (69 voted)

Reviews: 84% of readers found this page helpful

Author information

Name: Ray Christiansen

Birthday: 1998-05-04

Address: Apt. 814 34339 Sauer Islands, Hirtheville, GA 02446-8771

Phone: +337636892828

Job: Lead Hospitality Designer

Hobby: Urban exploration, Tai chi, Lockpicking, Fashion, Gunsmithing, Pottery, Geocaching

Introduction: My name is Ray Christiansen, I am a fair, good, cute, gentle, vast, glamorous, excited person who loves writing and wants to share my knowledge and understanding with you.