Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell ; 183(5): 1162-1184, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33242416

ABSTRACT

Research on astronaut health and model organisms have revealed six features of spaceflight biology that guide our current understanding of fundamental molecular changes that occur during space travel. The features include oxidative stress, DNA damage, mitochondrial dysregulation, epigenetic changes (including gene regulation), telomere length alterations, and microbiome shifts. Here we review the known hazards of human spaceflight, how spaceflight affects living systems through these six fundamental features, and the associated health risks of space exploration. We also discuss the essential issues related to the health and safety of astronauts involved in future missions, especially planned long-duration and Martian missions.


Subject(s)
Extraterrestrial Environment , Space Flight , Astronauts , Health , Humans , Microbiota , Risk Factors
3.
Int J Mol Sci ; 24(8)2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37108776

ABSTRACT

During space travel, astronauts will experience a unique environment that includes continuous exposure to microgravity and stressful living conditions. Physiological adaptation to this is a challenge and the effect of microgravity on organ development, architecture, and function is not well understood. How microgravity may impact the growth and development of an organ is an important issue, especially as space flight becomes more commonplace. In this work, we sought to address fundamental questions regarding microgravity using mouse mammary epithelial cells in 2D and 3D tissue cultures exposed to simulated microgravity. Mouse mammary HC11 cells contain a higher proportion of stem cells and were also used to investigate how simulated microgravity may impact mammary stem cell populations. In these studies, we exposed mouse mammary epithelial cells to simulated microgravity in 2D and then assayed for changes in cellular characteristics and damage levels. The microgravity treated cells were also cultured in 3D to form acini structures to define if simulated microgravity affects the cells' ability to organize correctly, a quality that is of key importance for mammary organ development. These studies identify changes occurring during exposure to microgravity that impact cellular characteristics such as cell size, cell cycle profiles, and levels of DNA damage. In addition, changes in the percentage of cells revealing various stem cell profiles were observed following simulated microgravity exposure. In summary, this work suggests microgravity may cause aberrant changes in mammary epithelial cells that lead to an increase in cancer risk.


Subject(s)
Space Flight , Weightlessness , Animals , Mice , Weightlessness/adverse effects , Cells, Cultured , Stem Cells , Epithelial Cells , Weightlessness Simulation
4.
Int J Mol Sci ; 20(3)2019 Feb 03.
Article in English | MEDLINE | ID: mdl-30717456

ABSTRACT

Space radiation has recently been considered a risk factor for astronauts' cardiac health. As an example, for the case of how to query and identify datasets within NASA's GeneLab database and demonstrate the database utility, we used an unbiased systems biology method for identifying key genes/drivers for the contribution of space radiation on the cardiovascular system. This knowledge can contribute to designing appropriate experiments targeting these specific pathways. Microarray data from cardiomyocytes of male C57BL/6 mice followed-up for 28 days after exposure to 900 mGy of 1 GeV proton or 150 mGy of 1 GeV/n 56Fe were compared to human endothelial cells (HUVECs) cultured for 7 days on the International Space Station (ISS). We observed common molecular pathways between simulated space radiation and HUVECs flown on the ISS. The analysis suggests FYN is the central driver/hub for the cardiovascular response to space radiation: the known oxidative stress induced immediately following radiation would only be transient and would upregulate FYN, which in turn would reduce reactive oxygen species (ROS) levels, protecting the cardiovascular system. The transcriptomic signature of exposure to protons was also much closer to the spaceflight signature than 56Fe's signature. To our knowledge, this is the first time GeneLab datasets were utilized to provide potential biological indications that the majority of ions on the ISS are protons, clearly illustrating the power of omics analysis. More generally, this work also demonstrates how to combine animal radiation studies done on the ground and spaceflight studies to evaluate human risk in space.


Subject(s)
Cardiovascular System/radiation effects , Myocytes, Cardiac/radiation effects , Proto-Oncogene Proteins c-fyn/genetics , Radiation, Ionizing , Space Flight , Transcriptome , Animals , Cardiovascular System/metabolism , Cells, Cultured , Cosmic Radiation , Gene Expression Regulation , Humans , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Oxidative Stress , Proto-Oncogene Proteins c-fyn/metabolism , Protons , Reactive Oxygen Species/metabolism
5.
Biomed Opt Express ; 15(4): 2561-2577, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38633084

ABSTRACT

To improve particle radiotherapy, we need a better understanding of the biology of radiation effects, particularly in heavy ion radiation therapy, where global responses are observed despite energy deposition in only a subset of cells. Here, we integrated a high-speed swept confocally-aligned planar excitation (SCAPE) microscope into a focused ion beam irradiation platform to allow real-time 3D structural and functional imaging of living biological samples during and after irradiation. We demonstrate dynamic imaging of the acute effects of irradiation on 3D cultures of U87 human glioblastoma cells, revealing characteristic changes in cellular movement and intracellular calcium signaling following ionizing irradiation.

6.
Nat Commun ; 15(1): 4825, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862542

ABSTRACT

Our previous research revealed a key microRNA signature that is associated with spaceflight that can be used as a biomarker and to develop countermeasure treatments to mitigate the damage caused by space radiation. Here, we expand on this work to determine the biological factors rescued by the countermeasure treatment. We performed RNA-sequencing and transcriptomic analysis on 3D microvessel cell cultures exposed to simulated deep space radiation (0.5 Gy of Galactic Cosmic Radiation) with and without the antagonists to three microRNAs: miR-16-5p, miR-125b-5p, and let-7a-5p (i.e., antagomirs). Significant reduction of inflammation and DNA double strand breaks (DSBs) activity and rescue of mitochondria functions are observed after antagomir treatment. Using data from astronaut participants in the NASA Twin Study, Inspiration4, and JAXA missions, we reveal the genes and pathways implicated in the action of these antagomirs are altered in humans. Our findings indicate a countermeasure strategy that can potentially be utilized by astronauts in spaceflight missions to mitigate space radiation damage.


Subject(s)
Astronauts , Cosmic Radiation , MicroRNAs , Space Flight , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Cosmic Radiation/adverse effects , DNA Breaks, Double-Stranded/radiation effects , Radiation Injuries/genetics , Radiation Injuries/prevention & control , Male , Mitochondria/radiation effects , Mitochondria/metabolism , Mitochondria/genetics , Female , Adult
7.
Microvasc Res ; 86: 11-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23220351

ABSTRACT

A limiting factor in the treatment of cancer with radiotherapy is the damage to surrounding normal tissue, particularly the vasculature. Vessel pathologies are a major feature of the side effects of radiotherapy and little is known about early events that could initiate subsequent diseases. We tested the hypothesis that gamma radiation has early damaging effects on the human endothelial barrier. Two models were used; Human Brain Microcapillary Endothelial Cells (HBMEC), and Human Umbilical Vein Endothelial Cells (HUVEC). Endpoints included Trans-Endothelial Electrical Resistance (TEER), barrier permeability to 10 kDa and 70 kDa tracer molecules, and the localization of F-actin, and junction proteins and the Platelet Endothelial Cell Adhesion Molecule (PECAM-1). Radiation induced a rapid and transient decrease in TEER at 3 h, with effects also seen at the radiotherapy doses. This dip in resistance correlated to the transient loss of PECAM-1 in discrete areas where cells often detached from the monolayer leaving gaps. Redistribution of PECAM-1 was also seen in 3-D human tissue models. By 6 h, the remaining cells had migrated to reseal the barrier, coincident with TEER returning to control levels. Resealed monolayers contained fewer cells per unit area and their barrier function was weakened as evidenced by an increased permeability over 24 h. This is the first demonstration of a transient and rapid effect of gamma radiation on human endothelial barriers that involves cell detachment and the loss of PECAM-1. Considering the association of cell adhesion molecules with vasculopathies, such an effect has the potential to be clinically relevant to the longer-term effects of radiotherapy.


Subject(s)
Capillary Permeability/radiation effects , Endothelium, Vascular/radiation effects , Gamma Rays/adverse effects , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Radiation Injuries/etiology , Actins/analysis , Cell Adhesion/radiation effects , Cell Adhesion Molecules/analysis , Cell Culture Techniques/methods , Cells, Cultured/chemistry , Cells, Cultured/physiology , Cells, Cultured/radiation effects , Cytoskeleton/ultrastructure , Electric Impedance , Endothelial Cells/chemistry , Endothelial Cells/physiology , Endothelial Cells/radiation effects , Endothelium, Vascular/chemistry , Endothelium, Vascular/physiology , Human Umbilical Vein Endothelial Cells/chemistry , Human Umbilical Vein Endothelial Cells/physiology , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Intercellular Junctions/radiation effects , Intercellular Junctions/ultrastructure , Microvessels/cytology , Organoids/chemistry , Organoids/physiology , Organoids/radiation effects
8.
Sci Transl Med ; 15(708): eabq1533, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37556555

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteins bind to host mitochondrial proteins, likely inhibiting oxidative phosphorylation (OXPHOS) and stimulating glycolysis. We analyzed mitochondrial gene expression in nasopharyngeal and autopsy tissues from patients with coronavirus disease 2019 (COVID-19). In nasopharyngeal samples with declining viral titers, the virus blocked the transcription of a subset of nuclear DNA (nDNA)-encoded mitochondrial OXPHOS genes, induced the expression of microRNA 2392, activated HIF-1α to induce glycolysis, and activated host immune defenses including the integrated stress response. In autopsy tissues from patients with COVID-19, SARS-CoV-2 was no longer present, and mitochondrial gene transcription had recovered in the lungs. However, nDNA mitochondrial gene expression remained suppressed in autopsy tissue from the heart and, to a lesser extent, kidney, and liver, whereas mitochondrial DNA transcription was induced and host-immune defense pathways were activated. During early SARS-CoV-2 infection of hamsters with peak lung viral load, mitochondrial gene expression in the lung was minimally perturbed but was down-regulated in the cerebellum and up-regulated in the striatum even though no SARS-CoV-2 was detected in the brain. During the mid-phase SARS-CoV-2 infection of mice, mitochondrial gene expression was starting to recover in mouse lungs. These data suggest that when the viral titer first peaks, there is a systemic host response followed by viral suppression of mitochondrial gene transcription and induction of glycolysis leading to the deployment of antiviral immune defenses. Even when the virus was cleared and lung mitochondrial function had recovered, mitochondrial function in the heart, kidney, liver, and lymph nodes remained impaired, potentially leading to severe COVID-19 pathology.


Subject(s)
COVID-19 , Cricetinae , Humans , Animals , Mice , COVID-19/pathology , SARS-CoV-2 , Rodentia , Genes, Mitochondrial , Lung/pathology
9.
Life Sci Space Res (Amst) ; 35: 105-112, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36336356

ABSTRACT

Future lunar missions and beyond will require new and innovative approaches to radiation countermeasures. The Translational Research Institute for Space Health (TRISH) is focused on identifying and supporting unique approaches to reduce risks to human health and performance on future missions beyond low Earth orbit. This paper will describe three funded and complementary avenues for reducing the risk to humans from radiation exposure experienced in deep space. The first focus is on identifying new therapeutic targets to reduce the damaging effects of radiation by focusing on high throughput genetic screens in accessible, sometimes called lower, organism models. The second focus is to design innovative approaches for countermeasure development with special attention to nucleotide-based methodologies that may constitute a more agile way to design therapeutics. The final focus is to develop new and innovative ways to test radiation countermeasures in a human model system. While animal studies continue to be beneficial in the study of space radiation, they can have imperfect translation to humans. The use of three-dimensional (3D) complex in vitro models is a promising approach to aid the development of new countermeasures and personalized assessments of radiation risks. These three distinct and unique approaches complement traditional space radiation efforts and should provide future space explorers with more options to safeguard their short and long-term health.


Subject(s)
Cosmic Radiation , Radiation Exposure , Radiation Protection , Space Flight , Animals , Humans , Cosmic Radiation/adverse effects , Radiation Protection/methods , Moon
10.
Cell Rep ; 37(3): 109839, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34624208

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation that have a major impact on many diseases and provide an exciting avenue toward antiviral therapeutics. From patient transcriptomic data, we determined that a circulating miRNA, miR-2392, is directly involved with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) machinery during host infection. Specifically, we show that miR-2392 is key in driving downstream suppression of mitochondrial gene expression, increasing inflammation, glycolysis, and hypoxia, as well as promoting many symptoms associated with coronavirus disease 2019 (COVID-19) infection. We demonstrate that miR-2392 is present in the blood and urine of patients positive for COVID-19 but is not present in patients negative for COVID-19. These findings indicate the potential for developing a minimally invasive COVID-19 detection method. Lastly, using in vitro human and in vivo hamster models, we design a miRNA-based antiviral therapeutic that targets miR-2392, significantly reduces SARS-CoV-2 viability in hamsters, and may potentially inhibit a COVID-19 disease state in humans.


Subject(s)
COVID-19/genetics , COVID-19/immunology , MicroRNAs/genetics , SARS-CoV-2/genetics , Adult , Aged , Aged, 80 and over , Animals , Antiviral Agents/pharmacology , Biomarkers/metabolism , Cricetinae , Female , Ferrets , Gene Expression Regulation , Glycolysis , Healthy Volunteers , Humans , Hypoxia , Inflammation , Male , Mice , Middle Aged , Proteomics/methods , ROC Curve , Rats , COVID-19 Drug Treatment
11.
bioRxiv ; 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-33948587

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation that have a major impact on many diseases and provides an exciting avenue towards antiviral therapeutics. From patient transcriptomic data, we have discovered a circulating miRNA, miR-2392, that is directly involved with SARS-CoV-2 machinery during host infection. Specifically, we show that miR-2392 is key in driving downstream suppression of mitochondrial gene expression, increasing inflammation, glycolysis, and hypoxia as well as promoting many symptoms associated with COVID-19 infection. We demonstrate miR-2392 is present in the blood and urine of COVID-19 positive patients, but not detected in COVID-19 negative patients. These findings indicate the potential for developing a novel, minimally invasive, COVID-19 detection method. Lastly, using in vitro human and in vivo hamster models, we have developed a novel miRNA-based antiviral therapeutic that targets miR-2392, significantly reduces SARS-CoV-2 viability in hamsters and may potentially inhibit a COVID-19 disease state in humans.

12.
Sci Rep ; 10(1): 2687, 2020 02 14.
Article in English | MEDLINE | ID: mdl-32060308

ABSTRACT

Radiotherapy combined with chemotherapy is the major treatment modality for human glioblastoma multiforme (GBM). GBMs eventually relapse after treatment and the average survival of GBM patients is less than two years. There is some evidence that cannabidiol (CBD) can induce cell death and increases the radiosensitivity of GBM by enhancing apoptosis. Beside initiation of death, CBD has been demonstrated as an inducer of autophagy. In the present study, we address the question whether CBD simultaneously induces a protective effect in GBM by upregulating autophagy. Addition of chloroquine that suppressed autophagic flux to 2D GBM cultures increased CBD-induced cell death, presenting proof for the protective autophagy. Blockage of autophagy upregulated radiation-induced cytotoxicity but only modestly affected the levels of cell death in CBD- or CBD/γ-irradiated 3D GBM cultures. Furthermore, CBD enhanced the pro-apoptotic activities of JNK1/2 and MAPK p38 signaling cascades while partially downregulated the pro-survival PI3K-AKT cascade, thereby changing a balance between cell death and survival. Suppression of JNK activation partially reduced CBD-induced cell death in 3D GBM cultures. In contrast, co-treatment of CBD-targeted cells with inhibitors of PI3K-AKT-NF-κB, IKK-NF-κB or JAK2-STAT3 pathways killed surviving GBM cells in both 2D and 3D cultures, potentially improving the therapeutic ratio of GBM.


Subject(s)
Autophagy/drug effects , Brain Neoplasms/drug therapy , Cannabidiol/pharmacology , Glioblastoma/drug therapy , Apoptosis/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Janus Kinase 2/genetics , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases/genetics , Radiation Tolerance/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/drug effects
13.
iScience ; 23(12): 101771, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33376971

ABSTRACT

Space radiation inhibits angiogenesis by two mechanisms depending on the linear energy transfer (LET). Using human 3D micro-vessel models, blockage of the early motile stage of angiogenesis was determined to occur after exposure to low LET ions (<3 KeV/AMU), whereas inhibition of the later stages occurs after exposure to high LET ions (>8 KeV/AMU). Strikingly, the combined effect is synergistic, detectible as low as 0.06 Gy making mixed ion space radiation more potent. Candidates for bystander transmission are microRNAs (miRNAs), and analysis on miRNA-seq data from irradiated mice shows that angiogenesis would in theory be downregulated. Further analysis of three previously identified miRNAs showed downregulation of their targets associated with angiogenesis and confirmed their involvement in angiogenesis pathways and increased health risks associated with cardiovascular disease. Finally, synthetic molecules (antagomirs) designed to inhibit the predicted miRNAs were successfully used to reverse the inhibition of angiogenesis.

14.
iScience ; 23(12): 101747, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33376970

ABSTRACT

Spaceflight missions can cause immune system dysfunction in astronauts with little understanding of immune outcomes in deep space. This study assessed immune responses in mice following ground-based, simulated deep spaceflight conditions, compared with data from astronauts on International Space Station missions. For ground studies, we simulated microgravity using the hindlimb unloaded mouse model alone or in combination with acute simulated galactic cosmic rays or solar particle events irradiation. Immune profiling results revealed unique immune diversity following each experimental condition, suggesting each stressor results in distinct circulating immune responses, with clear consequences for deep spaceflight. Circulating plasma microRNA sequence analysis revealed involvement in immune system dysregulation. Furthermore, a large astronaut cohort showed elevated inflammation during low-Earth orbit missions, thereby supporting our simulated ground experiments in mice. Herein, circulating immune biomarkers are defined by distinct deep space irradiation types coupled to simulated microgravity and could be targets for future space health initiatives.

15.
Int J Radiat Oncol Biol Phys ; 106(4): 857-866, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31759078

ABSTRACT

PURPOSE: The aim of this study is to characterize the effects of high-dose radiation therapy (HDRT) on Notch signaling components of the tumor vasculature. METHODS AND MATERIALS: Human umbilical vein endothelial cells monolayers were exposed to different single fraction doses of irradiation; ribonucleic acid RNA was isolated and polymerase chain reaction was performed for Notch signaling components. The vascular response to radiation therapy was examined in a xenograft model of neuroblastoma. Tumors were treated with 0 Gy, 2 Gy, and 12 Gy single fraction doses and analyzed by double immunofluorescence staining for Notch1, Notch ligands Jagged1 and Dll4, and the endothelial cell (EC) marker endomucin. To assess the role of Notch in vivo, NGP xenograft tumors expressing Fc or Notch1-1-24-decoy (a novel Notch inhibitor) were treated with 0 Gy and 12 Gy. Immunofluorescence staining for endomucin and endomucin/αSMA was performed to analyze the effect of combination treatment on tumor EC and endothelial-to-mesenchymal-transition (EndMT), respectively. RESULTS: In human umbilical vein endothelial cells monolayers doses ≥8 Gy increased expression of NOTCH1, JAG1, and Notch target genes HEY1 and HEY2 as early as 6 hours after irradiation. In vivo, 12 Gy significantly increased Notch1 and Jagged1 in tumor ECs compared with 0 Gy or 2 Gy after 72 hours. Combining HDRT with Notch inhibition using the Notch1-1-24-decoy resulted in a greater loss of EC coverage of tumor vessels than HDRT alone at 6 hours and 72 hours post treatment. Notch inhibition reduced EndMT induced by HDRT, as indicated by diminished αSMA staining in ECs. CONCLUSIONS: HDRT induced Notch1 expression and increased Notch1 signaling in the endothelial component of tumor vasculature, which was not observed with lower doses. This increase in Notch1 activation might protect tumor vessels from HDRT induced damage and regulate EndMT process.


Subject(s)
Neovascularization, Pathologic/metabolism , Radiation Dosage , Receptor, Notch1/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic , Epithelial-Mesenchymal Transition/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Jagged-1 Protein/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , Radiotherapy Dosage , Repressor Proteins/metabolism , Signal Transduction/radiation effects
16.
Cell Rep ; 33(10): 108448, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33242410

ABSTRACT

We have identified and validated a spaceflight-associated microRNA (miRNA) signature that is shared by rodents and humans in response to simulated, short-duration and long-duration spaceflight. Previous studies have identified miRNAs that regulate rodent responses to spaceflight in low-Earth orbit, and we have confirmed the expression of these proposed spaceflight-associated miRNAs in rodents reacting to simulated spaceflight conditions. Moreover, astronaut samples from the NASA Twins Study confirmed these expression signatures in miRNA sequencing, single-cell RNA sequencing (scRNA-seq), and single-cell assay for transposase accessible chromatin (scATAC-seq) data. Additionally, a subset of these miRNAs (miR-125, miR-16, and let-7a) was found to regulate vascular damage caused by simulated deep space radiation. To demonstrate the physiological relevance of key spaceflight-associated miRNAs, we utilized antagomirs to inhibit their expression and successfully rescue simulated deep-space-radiation-mediated damage in human 3D vascular constructs.


Subject(s)
Circulating MicroRNA/genetics , MicroRNAs/genetics , Weightlessness/adverse effects , Animals , Female , Gene Expression , Gene Expression Profiling/methods , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Rats , Sequence Analysis, RNA/methods , Space Flight , Transcriptome/genetics , Weightlessness Simulation/methods
18.
J Neurosci ; 27(21): 5823-34, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17522326

ABSTRACT

Recent evidence has implicated dynein and its regulatory factors dynactin and LIS1 in neuronal and non-neuronal cell migration. In the current study we sought to test whether effects on neuronal cell motility might reflect, in part, a role for these proteins in the growth cone. In chick sensory neurons subjected to acute laminin treatment dynein, dynactin, and LIS1 were mobilized strikingly and rapidly to the leading edge of the growth cone, where they were seen to be associated with microtubules converging into the laminin-induced axonal outgrowths. To interfere acutely with LIS1 and dynein function and to minimize secondary phenotypic effects, we injected antibodies to these proteins just before axon initiation. Antibody to both proteins produced an almost complete block of laminin-induced growth cone remodeling and the underlying reorganization of microtubules. Penetration of microtubules into the peripheral zone of differentiating axonal growth cones was decreased dramatically by antibody injection, as judged by live analysis of enhanced green fluorescent protein-tubulin and the microtubule tip-associated EB3 (end-binding protein 3). Dynein and LIS1 inhibition had no detectable effect on microtubule assembly but reduced the ability of microtubules to resist retrograde actin flow. In hippocampal neurons dynein, dynactin, and LIS1 were enriched in axonal growth cones at stage 3, and both growth cone organization and axon elongation were altered by LIS1 RNA interference. Together, our data indicate that dynein and LIS1 play a surprisingly prominent role in microtubule advance during growth cone remodeling associated with axonogenesis. These data may explain, in part, the role of these proteins in brain developmental disease and support an important role in diverse aspects of neuronal differentiation and nervous system development.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Axons/physiology , Cytoplasm/physiology , Dyneins/physiology , Growth Cones/physiology , Microtubule-Associated Proteins/physiology , Microtubules/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/analysis , Animals , Axons/chemistry , Chick Embryo , Cytoplasm/chemistry , Dyneins/analysis , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/physiology , Growth Cones/chemistry , Microtubule-Associated Proteins/analysis , Microtubules/chemistry , Rats
19.
Radiat Res ; 186(6): 602-613, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27905868

ABSTRACT

A significant target for radiation-induced effects is the microvascular system, which is critical to healthy tissue function and its pathology is linked to disrupted endothelial barrier function. Low-linear energy transfer (LET) ionizing radiation is a source of noncancer pathologies in humans and little is known about the early events that could initiate subsequent diseases. However, it is well known that gamma radiation causes a very early disruption of the endothelial barrier at doses below those required for cytotoxic effects. After irradiation of human umbilical vein endothelial cells (HUVECs) to doses as low as 2 Gy, transendothelial electrical resistance (TEER) is transiently reduced at 3 h, and the platelet-derived endothothelial cell adhesion molecule (PECAM-1 or CD31) is uncoupled from the cells along with the release of endothelial microparticles (EMPs). In this study, we measured TEER reduction as an indicator of barrier function loss, and specifically examined the shedding of EMPs from human endothelial barrier models after a variety of low-LET irradiations, including photons and charged particles. Our findings showed two TEER responses, dependent on radiation type and environmental conditions. The first response was diminishing oscillations of TEER, which occurred during the first 10 h postirradiation. This response occurred after a 5 Gy proton or helium-ion (1 GeV/n) dose in addition to a 5 Gy gamma or X radiation dose. This occurred only in the presence of multiple growth factors and did not show a dose response, nor was it associated with EMP release. The second response was a single acute drop in TEER at 3 h after photon irradiation. Dose response was observed and was associated with the shedding of EMPs in 2D barrier cultures and in 3D vessel models. In this case, helium-ion and proton irradiations did not induce a drop in TEER or shedding of EMPs. The photon radiation effects was observed both in serum-free media and in the presence of multiple growth factors, indicating that it occurs under a range of environmental conditions. These results show an acute response of the human endothelial barrier that is relevant to photon irradiation. Significantly, it involves the release of EMPs, which have recently attracted attention due to their emerging clinical importance.


Subject(s)
Cell-Derived Microparticles/radiation effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/radiation effects , Linear Energy Transfer , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Cell-Derived Microparticles/metabolism , Dose-Response Relationship, Radiation , Gamma Rays , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Protein Transport/radiation effects , Protons , Serum/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL