Micro-RNA and Proteomic Profiles of Plasma-Derived Exosomes from Irradiated Mice Reveal Molecular Changes Preventing Apoptosis in Neonatal Cerebellum

Cell communication via exosomes is capable of influencing cell fate in stress situations such as exposure to ionizing radiation. In vitro and in vivo studies have shown that exosomes might play a role in out-of-target radiation effects by carrying molecular signaling mediators of radiation damage, as well as opposite protective functions resulting in resistance to radiotherapy. However, a global understanding of exosomes and their radiation-induced regulation, especially within the context of an intact mammalian organism, has been lacking. In this in vivo study, we demonstrate that, compared to sham-irradiated (SI) mice, a distinct pattern of proteins and miRNAs is found packaged into circulating plasma exosomes after whole-body and partial-body irradiation (WBI and PBI) with 2 Gy X-rays. A high number of deregulated proteins (59% of WBI and 67% of PBI) was found in the exosomes of irradiated mice. In total, 57 and 13 miRNAs were deregulated in WBI and PBI groups, respectively, suggesting that the miRNA cargo is influenced by the tissue volume exposed to radiation. In addition, five miRNAs (miR-99b-3p, miR-200a-3p, miR-200a, miR-182-5p, miR-182) were commonly overexpressed in the exosomes from the WBI and PBI groups. In this study, particular emphasis was also given to the determination of the in vivo effect of exosome transfer by intracranial injection in the highly radiosensitive neonatal cerebellum at postnatal day 3. In accordance with a major overall anti-apoptotic function of the commonly deregulated miRNAs, here, we report that exosomes from the plasma of irradiated mice, especially in the case of WBI, prevent radiation-induced apoptosis, thus holding promise for exosome-based future therapeutic applications against radiation injury.

[1]  Haodong Lin,et al.  MicroRNA-182 improves spinal cord injury in mice by modulating apoptosis and the inflammatory response via IKKβ/NF-κB , 2021, Laboratory Investigation.

[2]  K. N. Bhanu Prakash,et al.  Early postnatal irradiation‐induced age‐dependent changes in adult mouse brain: MRI based characterization , 2021, BMC neuroscience.

[3]  S. Pazzaglia,et al.  miRNA-Signature of Irradiated Ptch1+/– Mouse Lens is Dependent on Genetic Background , 2021, Radiation Research.

[4]  Damien Traynor,et al.  Out-of-Field Hippocampus from Partial-Body Irradiated Mice Displays Changes in Multi-Omics Profile and Defects in Neurogenesis , 2021, International journal of molecular sciences.

[5]  P. Gong,et al.  Plasma-Derived Exosomes Boost the Healing of Irradiated Wound by Regulating Cell Proliferation and Ferroptosis. , 2021, Journal of biomedical nanotechnology.

[6]  Qian Liu,et al.  miR-182-5p contributes to radioresistance in nasopharyngeal carcinoma by regulating BNIP3 expression , 2020, Molecular medicine reports.

[7]  M. Germain,et al.  Selective packaging of mitochondrial proteins into extracellular vesicles prevents the release of mitochondrial DAMPs , 2020, Nature Communications.

[8]  D. Sinclair,et al.  Extracellular Vesicles for the Treatment of Radiation-Induced Normal Tissue Toxicity in the Lung , 2021, Frontiers in Oncology.

[9]  Kexiang Liu,et al.  The Role of Exosomes and Exosomal MicroRNA in Cardiovascular Disease , 2021, Frontiers in Cell and Developmental Biology.

[10]  Lihua Dong,et al.  Mesenchymal Stem Cell-Derived Exosomes: Biological Function and Their Therapeutic Potential in Radiation Damage , 2020, Cells.

[11]  Damien Traynor,et al.  Phenotypic and Functional Characteristics of Exosomes Derived from Irradiated Mouse Organs and Their Role in the Mechanisms Driving Non-Targeted Effects , 2020, International journal of molecular sciences.

[12]  D. Strunk,et al.  Functional assays to assess the therapeutic potential of extracellular vesicles , 2020, Journal of extracellular vesicles.

[13]  S. Du,et al.  Exosome: A Review of Its Classification, Isolation Techniques, Storage, Diagnostic and Targeted Therapy Applications , 2020, International journal of nanomedicine.

[14]  Katarzyna Jonak,et al.  The function of miR-200 family in oxidative stress response evoked in cancer chemotherapy and radiotherapy. , 2020, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[15]  W. Ngwa,et al.  Single Radiotherapy Fraction with Local Anti-CD40 Therapy Generates Effective Abscopal Responses in Mouse Models of Cervical Cancer , 2020, Cancers.

[16]  S. Tapio,et al.  Radiation Exposure of Peripheral Mononuclear Blood Cells Alters the Composition and Function of Secreted Extracellular Vesicles , 2020, International journal of molecular sciences.

[17]  Gang Chen,et al.  Exosome and Secretion: Action On? , 2020, Advances in experimental medicine and biology.

[18]  D. Hladik,et al.  CREB Signaling Mediates Dose-dependent Radiation Response in the Murine Hippocampus Two Years after Total Body Exposure. , 2019, Journal of proteome research.

[19]  M. C. Angulo,et al.  Functional equivalence of stem cell and stem cell‐derived extracellular vesicle transplantation to repair the irradiated brain , 2019, Stem cells translational medicine.

[20]  J. Lötvall,et al.  Mitochondrial protein enriched extracellular vesicles discovered in human melanoma tissues can be detected in patient plasma , 2019, Journal of extracellular vesicles.

[21]  André F. Rendeiro,et al.  Mitochondria Are a Subset of Extracellular Vesicles Released by Activated Monocytes and Induce Type I IFN and TNF Responses in Endothelial Cells. , 2019, Circulation research.

[22]  Gang Chen,et al.  MiR‐182 enhances radioresistance in non‐small cell lung cancer cells by regulating FOXO3 , 2019, Clinical and experimental pharmacology & physiology.

[23]  Martin Eisenacher,et al.  The PRIDE database and related tools and resources in 2019: improving support for quantification data , 2018, Nucleic Acids Res..

[24]  A. Cheema,et al.  Plasma Derived Exosomal Biomarkers of Exposure to Ionizing Radiation in Nonhuman Primates , 2018, International journal of molecular sciences.

[25]  J. Ravanat,et al.  Targeted and Off-Target (Bystander and Abscopal) Effects of Radiation Therapy: Redox Mechanisms and Risk/Benefit Analysis , 2018, Antioxidants & redox signaling.

[26]  Tushar Patel,et al.  Circulating Extracellular Vesicles in Human Disease. , 2018, The New England journal of medicine.

[27]  D. Molin,et al.  Extracellular Vesicles Work as a Functional Inflammatory Mediator Between Vascular Endothelial Cells and Immune Cells , 2018, Front. Immunol..

[28]  P. Seth,et al.  Decorin-Modified Umbilical Cord Mesenchymal Stem Cells (MSCs) Attenuate Radiation-Induced Lung Injuries via Regulating Inflammation, Fibrotic Factors, and Immune Responses. , 2018, International journal of radiation oncology, biology, physics.

[29]  L. Lannfelt,et al.  Alzheimer’s disease pathology propagation by exosomes containing toxic amyloid-beta oligomers , 2018, Acta Neuropathologica.

[30]  A. Buchan,et al.  Circulating endothelial cell-derived extracellular vesicles mediate the acute phase response and sickness behaviour associated with CNS inflammation , 2017, Scientific Reports.

[31]  Wangsheng Wang,et al.  Induction of pro-inflammatory genes by serum amyloid A1 in human amnion fibroblasts , 2017, Scientific Reports.

[32]  R. Cole,et al.  Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions , 2017, Science Signaling.

[33]  S. Thom,et al.  Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury , 2017, Journal of Neuroinflammation.

[34]  Li Kong,et al.  Abscopal effect of metastatic pancreatic cancer after local radiotherapy and granulocyte-macrophage colony-stimulating factor therapy , 2017, Cancer biology & therapy.

[35]  C. McDonald,et al.  Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours , 2017, Nature Reviews Neurology.

[36]  F. Fan,et al.  Mesenchymal stem cells stimulate intestinal stem cells to repair radiation-induced intestinal injury , 2016, Cell Death and Disease.

[37]  P. Widłak,et al.  The Influence of Ionizing Radiation on Exosome Composition, Secretion and Intercellular Communication , 2016, Protein and peptide letters.

[38]  M. Schachner,et al.  Improvement of neuronal cell survival by astrocyte‐derived exosomes under hypoxic and ischemic conditions depends on prion protein , 2016, Glia.

[39]  L. Ramenghi,et al.  Exosomes from human mesenchymal stem cells conduct aerobic metabolism in term and preterm newborn infants , 2016, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[40]  R. Yentrapalli,et al.  Exosomes Derived from Squamous Head and Neck Cancer Promote Cell Survival after Ionizing Radiation , 2016, PloS one.

[41]  F. Wendler,et al.  Extracellular vesicles round off communication in the nervous system , 2016, Nature Reviews Neuroscience.

[42]  C. Théry,et al.  Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes , 2016, Proceedings of the National Academy of Sciences.

[43]  M. Kadhim,et al.  Exosome-Mediated Telomere Instability in Human Breast Epithelial Cancer Cells after X Irradiation , 2016, Radiation Research.

[44]  K. N. Yu,et al.  Modulation of NF-κB in rescued irradiated cells. , 2015, Radiation protection dosimetry.

[45]  Bing Wang,et al.  Exosome-mediated microRNA transfer plays a role in radiation-induced bystander effect , 2015, RNA biology.

[46]  J. Kurie,et al.  BMP4 depletion by miR-200 inhibits tumorigenesis and metastasis of lung adenocarcinoma cells , 2015, Molecular Cancer.

[47]  S. Hauck,et al.  The Proteome of Native Adult Müller Glial Cells From Murine Retina* , 2015, Molecular & Cellular Proteomics.

[48]  Yan Zhang,et al.  Therapeutic Potential of Umbilical Cord Mesenchymal Stem Cells for Inhibiting Myofibroblastic Differentiation of Irradiated Human Lung Fibroblasts. , 2015, The Tohoku journal of experimental medicine.

[49]  A. Hill,et al.  Extracellular vesicles--Their role in the packaging and spread of misfolded proteins associated with neurodegenerative diseases. , 2015, Seminars in cell & developmental biology.

[50]  M. Kadhim,et al.  The non-targeted effects of radiation are perpetuated by exosomes. , 2015, Mutation research.

[51]  Brock A. Humphries,et al.  The microRNA-200 family: small molecules with novel roles in cancer development, progression and therapy , 2015, Oncotarget.

[52]  L. O’Driscoll,et al.  Biological properties of extracellular vesicles and their physiological functions , 2015, Journal of extracellular vesicles.

[53]  F. Sánchez‐Madrid,et al.  Sorting it out: regulation of exosome loading. , 2014, Seminars in cancer biology.

[54]  H. Luhmann,et al.  Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation , 2014, Philosophical Transactions of the Royal Society B: Biological Sciences.

[55]  Yankai Xia,et al.  Decreased MiR-200a/141 Suppress Cell Migration and Proliferation by Targeting PTEN in Hirschsprung's Disease , 2014, Cellular Physiology and Biochemistry.

[56]  C. Bracken,et al.  MiR-200 can repress breast cancer metastasis through ZEB1-independent but moesin-dependent pathways , 2014, Oncogene.

[57]  K. Mizuguchi,et al.  Blockade of TLR3 protects mice from lethal radiation-induced gastrointestinal syndrome , 2014, Nature Communications.

[58]  Concha Gil,et al.  General statistical framework for quantitative proteomics by stable isotope labeling. , 2014, Journal of proteome research.

[59]  K. Camphausen,et al.  Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration. , 2013, Translational oncology.

[60]  Aiman S Saab,et al.  Neurotransmitter-Triggered Transfer of Exosomes Mediates Oligodendrocyte–Neuron Communication , 2013, PLoS biology.

[61]  J. Lin,et al.  A critical role of toll-like receptor 4 (TLR4) and its' in vivo ligands in basal radio-resistance , 2013, Cell Death and Disease.

[62]  Mariateresa Mancuso,et al.  Dose and spatial effects in long-distance radiation signaling in vivo: implications for abscopal tumorigenesis. , 2013, International journal of radiation oncology, biology, physics.

[63]  H. Northoff,et al.  Mesenchymal stromal/stem cells markers in the human bone marrow. , 2013, Cytotherapy.

[64]  J. Lan,et al.  Gene-modified mesenchymal stem cells protect against radiation-induced lung injury. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[65]  Bernhard Mlecnik,et al.  CluePedia Cytoscape plugin: pathway insights using integrated experimental and in silico data , 2013, Bioinform..

[66]  C. Verfaillie,et al.  Immunological characteristics of human mesenchymal stem cells and multipotent adult progenitor cells , 2013, Immunology and cell biology.

[67]  Anindya Dutta,et al.  The miR-99 family regulates the DNA damage response through its target SNF2H , 2012, Oncogene.

[68]  S. Pazzaglia,et al.  The radiation bystander effect and its potential implications for human health. , 2012, Current molecular medicine.

[69]  Jason C. Young,et al.  Prevention and Mitigation of Acute Radiation Syndrome in Mice by Synthetic Lipopeptide Agonists of Toll-Like Receptor 2 (TLR2) , 2012, PloS one.

[70]  M. Kadhim,et al.  Possible Role of Exosomes Containing RNA in Mediating Nontargeted Effect of Ionizing Radiation , 2012, Radiation research.

[71]  Simon C Watkins,et al.  Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes. , 2012, Blood.

[72]  Dong Yu,et al.  TLR9 Agonist Protects Mice from Radiation-Induced Gastrointestinal Syndrome , 2012, PloS one.

[73]  S. Pazzaglia,et al.  Role of connexin43 and ATP in long-range bystander radiation damage and oncogenesis in vivo , 2011, Oncogene.

[74]  P. Fasanaro,et al.  miR-200c is upregulated by oxidative stress and induces endothelial cell apoptosis and senescence via ZEB1 inhibition , 2011, Cell Death and Differentiation.

[75]  J. Privratsky,et al.  PECAM-1: conflicts of interest in inflammation. , 2010, Life sciences.

[76]  K. Midwood,et al.  DAMPening Inflammation by Modulating TLR Signalling , 2010, Mediators of inflammation.

[77]  Richard J. Simpson,et al.  Proteomics Analysis of A33 Immunoaffinity-purified Exosomes Released from the Human Colon Tumor Cell Line LIM1215 Reveals a Tissue-specific Protein Signature* , 2009, Molecular & Cellular Proteomics.

[78]  Robert L Moritz,et al.  Exosomes: proteomic insights and diagnostic potential , 2009, Expert review of proteomics.

[79]  M. Mann,et al.  Universal sample preparation method for proteome analysis , 2009, Nature Methods.

[80]  Michele Guescini,et al.  Astrocytes and Glioblastoma cells release exosomes carrying mtDNA , 2009, Journal of Neural Transmission.

[81]  Mikael Olsson String , 2020, Encyclopedia of Algorithms.

[82]  M. Pimpinella,et al.  Oncogenic bystander radiation effects in Patched heterozygous mouse cerebellum , 2008, Proceedings of the National Academy of Sciences.

[83]  Joseph A. DiDonato,et al.  An Agonist of Toll-Like Receptor 5 Has Radioprotective Activity in Mouse and Primate Models , 2008, Science.

[84]  J. Lötvall,et al.  Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells , 2007, Nature Cell Biology.

[85]  C. Mothersill,et al.  The Involvement of Calcium and MAP Kinase Signaling Pathways in the Production of Radiation-Induced Bystander Effects , 2006, Radiation research.

[86]  S. Pazzaglia,et al.  Linking DNA damage to medulloblastoma tumorigenesis in patched heterozygous knockout mice , 2006, Oncogene.

[87]  M. Heyman,et al.  Phenotypic and functional characterization of intestinal epithelial exosomes. , 2005, Blood cells, molecules & diseases.

[88]  A. Mee,et al.  Diagnostic radiation exposure and cancer risk , 2005, Gut.

[89]  Kiheung Kim,et al.  Ko Kuei Chen: a pioneer of modern pharmacological research in China , 2022, Protein & cell.

[90]  Kevin M Prise,et al.  Targeted cytoplasmic irradiation induces bystander responses. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[91]  G. Raposo,et al.  Exosomes: endosomal-derived vesicles shipping extracellular messages. , 2004, Current opinion in cell biology.

[92]  W. Faigle,et al.  Cells release prions in association with exosomes. , 2004, Proceedings of the National Academy of Sciences of the United States of America.

[93]  M. Bittner,et al.  Induction of Gene Expression as a Monitor of Exposure to Ionizing Radiation , 2001, Radiation research.

[94]  Laurence Zitvogel,et al.  Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming , 2001, Nature Medicine.

[95]  P Lambin,et al.  Low-dose hypersensitivity: current status and possible mechanisms. , 2001, International journal of radiation oncology, biology, physics.

[96]  B. Lehnert,et al.  Effects of ionizing radiation in targeted and nontargeted cells. , 2000, Archives of biochemistry and biophysics.

[97]  E. Wright Inducible genomic instability: Inducible genomic instability: New insights into the biological effects of ionizing radiation , 2000, Medicine, conflict, and survival.

[98]  H. Geuze,et al.  Selective Enrichment of Tetraspan Proteins on the Internal Vesicles of Multivesicular Endosomes and on Exosomes Secreted by Human B-lymphocytes* , 1998, The Journal of Biological Chemistry.

[99]  S. Wolff The adaptive response in radiobiology: evolving insights and implications. , 1998, Environmental health perspectives.

[100]  E. Wright Radiation-induced genomic instability in haemopoietic cells. , 1998, International journal of radiation biology.

[101]  Proceedings of the American Statistical Association Conference on Radiation and Health. Radiation Risk and Interactions. South Carolina, June 28-July 2, 1992. Abstracts. , 1993, Radiation research.