Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 499
Filter
1.
Lasers Med Sci ; 39(1): 122, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38703271

ABSTRACT

Pulsed dye lasers are used effectively in the treatment of psoriasis with long remission time and limited side effects. It is, however, not completely understood which biological processes underlie its favorable outcome. Pulsed dye laser treatment at 585-595 nm targets hemoglobin in the blood, inducing local hyperthermia in surrounding blood vessels and adjacent tissues. While the impact of destructive temperatures on blood vessels has been well studied, the effects of lower temperatures on the function of several cell types within the blood vessel wall and its periphery are not known. The aim of our study is to assess the functionality of isolated blood vessels after exposure to moderate hyperthermia (45 to 60°C) by evaluating the function of endothelial cells, smooth muscle cells, and vascular nerves. We measured blood vessel functionality of rat mesenteric arteries (n=19) by measuring vascular contraction and relaxation before and after heating vessels in a wire myograph. To this end, we elicited vascular contraction by addition of either high potassium solution or the thromboxane analogue U46619 to stimulate smooth muscle cells, and electrical field stimulation (EFS) to stimulate nerves. For measurement of endothelium-dependent relaxation, we used methacholine. Each vessel was exposed to one temperature in the range of 45-60°C for 30 seconds and a relative change in functional response after hyperthermia was determined by comparison with the response per stimulus before heating. Non-linear regression was used to fit our dataset to obtain the temperature needed to reduce blood vessel function by 50% (Half maximal effective temperature, ET50). Our findings demonstrate a substantial decrease in relative functional response for all three cell types following exposure to 55°C-60°C. There was no significant difference between the ET50 values of the different cell types, which was between 55.9°C and 56.9°C (P>0.05). Our data show that blood vessel functionality decreases significantly when exposed to temperatures between 55°C-60°C for 30 seconds. The results show functionality of endothelial cells, smooth muscle cells, and vascular nerves is similarly impaired. These results help to understand the biological effects of hyperthermia and may aid in tailoring laser and light strategies for selective photothermolysis that contribute to disease modification of psoriasis after pulsed dye laser treatment.


Subject(s)
Lasers, Dye , Animals , Rats , Male , Lasers, Dye/therapeutic use , Myocytes, Smooth Muscle/physiology , Myocytes, Smooth Muscle/radiation effects , Vasodilation/radiation effects , Vasodilation/physiology , Temperature , Muscle, Smooth, Vascular/radiation effects , Muscle, Smooth, Vascular/physiology , Endothelial Cells/radiation effects , Endothelial Cells/physiology , Vasoconstriction/radiation effects , Vasoconstriction/physiology , Endothelium, Vascular/radiation effects , Rats, Wistar
2.
Aging (Albany NY) ; 16(8): 6673-6693, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38683123

ABSTRACT

PURPOSE: The objective of this study was to investigate the senescent phenotypes of human corneal endothelial cells (hCEnCs) upon treatment with ultraviolet (UV)-A. METHODS: We assessed cell morphology, senescence-associated ß-galactosidase (SA-ß-gal) activity, cell proliferation and expression of senescence markers (p16 and p21) in hCEnCs exposed to UV-A radiation, and senescent hCEnCs induced by ionizing radiation (IR) were used as positive controls. We performed RNA sequencing and proteomics analyses to compare gene and protein expression profiles between UV-A- and IR-induced senescent hCEnCs, and we also compared the results to non-senescent hCEnCs. RESULTS: Cells exposed to 5 J/cm2 of UV-A or to IR exhibited typical senescent phenotypes, including enlargement, increased SA-ß-gal activity, decreased cell proliferation and elevated expression of p16 and p21. RNA-Seq analysis revealed that 83.9% of the genes significantly upregulated and 82.6% of the genes significantly downregulated in UV-A-induced senescent hCEnCs overlapped with the genes regulated in IR-induced senescent hCEnCs. Proteomics also revealed that 93.8% of the proteins significantly upregulated in UV-A-induced senescent hCEnCs overlapped with those induced by IR. In proteomics analyses, senescent hCEnCs induced by UV-A exhibited elevated expression levels of several factors part of the senescence-associated secretory phenotype. CONCLUSIONS: In this study, where senescence was induced by UV-A, a more physiological stress for hCEnCs compared to IR, we determined that UV-A modulated the expression of many genes and proteins typically altered upon IR treatment, a more conventional method of senescence induction, even though UV-A also modulated specific pathways unrelated to IR.


Subject(s)
Cell Proliferation , Cellular Senescence , Endothelial Cells , Ultraviolet Rays , Humans , Cellular Senescence/radiation effects , Ultraviolet Rays/adverse effects , Cell Proliferation/radiation effects , Endothelial Cells/radiation effects , Endothelial Cells/metabolism , Endothelium, Corneal/radiation effects , Endothelium, Corneal/metabolism , Cells, Cultured , Proteomics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , beta-Galactosidase/metabolism , beta-Galactosidase/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics
3.
Int J Radiat Biol ; 100(5): 756-766, 2024.
Article in English | MEDLINE | ID: mdl-38489594

ABSTRACT

PURPOSE: People are exposed to low-dose radiation in medical diagnosis, occupational, or life circumstances, but the effect of low-dose radiation on human health is still controversial. The biological effects of radiation below 100 mGy are still unproven. In this study, we observed the effects of low-dose radiation (100 mGy) on gene expression in human coronary artery endothelial cells (HCAECs) and its effect on molecular signaling. MATERIALS AND METHODS: HCAECs were exposed to 100 mGy ionizing radiation at 6 mGy/h (low-dose-rate) or 288 mGy/h (high-dose-rate). After 72 h, total RNA was extracted from sham or irradiated cells for Quant-Seq 3'mRNA-Seq, and bioinformatic analyses were performed using Metascape. Gene profiling was validated using qPCR. RESULTS: Compared to the non-irradiated control group, 100 mGy of ionizing radiation at 6 mGy/h altered the expression of 194 genes involved in signaling pathways related to heart contraction, blood circulation, and cardiac myofibril assembly differentially. However, 100 mGy at 288 mGy/h altered expression of 450 genes involved in cell cycle-related signaling pathways, including cell division, nuclear division, and mitosis differentially. Additionally, gene signatures responding to low-dose radiation, including radiation dose-specific gene profiles (HIST1H2AI, RAVER1, and POTEI) and dose-rate-specific gene profiles (MYL2 for the low-dose-rate and DHRS9 and CA14 for the high-dose-rate) were also identified. CONCLUSIONS: We demonstrated that 100 mGy low-dose radiation could alter gene expression and molecular signaling pathways at the low-dose-rate and the high-dose-rate differently. Our findings provide evidence for further research on the potential impact of low-dose radiation on cardiovascular function.


Subject(s)
Computational Biology , Coronary Vessels , Dose-Response Relationship, Radiation , Endothelial Cells , Transcriptome , Humans , Coronary Vessels/radiation effects , Coronary Vessels/cytology , Endothelial Cells/radiation effects , Endothelial Cells/metabolism , Transcriptome/radiation effects , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Radiation Dosage , Signal Transduction/radiation effects
4.
Sci Prog ; 107(1): 368504241228668, 2024.
Article in English | MEDLINE | ID: mdl-38385346

ABSTRACT

OBJECTIVE: This study aimed to investigate the effects of low-dose radiation on the abdominal aorta of mice and vascular endothelial cells. METHODS: Wild-type and tumor-bearing mice were exposed to 15 sessions of low-dose irradiation, resulting in cumulative radiation doses of 187.5, 375, and 750 mGy. The effect on the cardiovascular system was assessed. Immunohistochemistry analyzed protein expressions of PAPP-A, CD62, P65, and COX-2 in the abdominal aorta. Microarray technology, Gene Ontology analysis, and pathway enrichment analysis evaluated gene expression changes in endothelial cells exposed to 375 mGy X-ray. Cell viability was assessed using the Cell Counting Kit 8 assay. Immunofluorescence staining measured γ-H2AX levels, and real-time polymerase chain reaction quantified mRNA levels of interleukin-6 (IL-6), ICAM-1, and Cx43. RESULTS: Hematoxylin and eosin staining revealed thickening of the inner membranes and irregular arrangement of smooth muscle cells in the media membrane at 375 and 750 mGy. Inflammation was observed in the inner membranes at 750 mGy, with a clear inflammatory response in the hearts of tumor-bearing mice. Immunohistochemistry indicated increased levels of PAPP-A, P65, and COX-2 post-irradiation. Microarray analysis showed 425 up-regulated and 235 down-regulated genes, associated with processes like endothelial cell-cell adhesion, IL-6, and NF-κB signaling. Cell Counting Kit 8 assay results indicated inhibited viability at 750 mGy in EA.hy926 cells. Immunofluorescence staining demonstrated a dose-dependent increase in γ-H2AX foci. Reverse transcription quantitative PCR results showed increased expression of IL6, ICAM-1, and Cx43 in EA.hy926 cells post 750 mGy X-ray exposure. CONCLUSION: Repeated low-dose ionizing radiation exposures triggered the development of pro-atherosclerotic phenotypes in mice and damage to vascular endothelial cells.


Subject(s)
Endothelial Cells , Neoplasms , Humans , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Intercellular Adhesion Molecule-1/metabolism , Connexin 43/genetics , Interleukin-6/genetics , Cyclooxygenase 2/genetics , Pregnancy-Associated Plasma Protein-A , Radiation, Ionizing , Phenotype
5.
Int J Radiat Oncol Biol Phys ; 117(1): 223-236, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37059236

ABSTRACT

PURPOSE: Radiation therapy (RT) significantly increased the incidence of coronary artery diseases, especially atherosclerosis. Endothelial dysfunction has been the major side effect of RT among tumor patients who received RT. However, the involvement between endothelial dysfunction and radiation-induced atherosclerosis (RIA) remains unclear. Here, we constructed a murine model of RIA, aiming to uncover its underlying mechanisms and identify novel strategies for RIA prevention and treatment. METHODS AND MATERIALS: Eight-week-old ApoE-/- mice that were fed a Western diet were subjected to partial carotid ligation (PCL). Four weeks later, ionizing radiation (IR) of 10 Gy was performed to verify the detrimental role of IR on atherogenesis. Ultrasound imaging, RT quantitative polymerase chain reaction, histopathology and immunofluorescence, and biochemical analysis were performed 4 weeks after IR. To study the involvement of endothelial ferroptosis induced by IR in RIA, mice after IR were administrated with ferroptosis agonist (cisplatin) or antagonist (ferrostatin-1) intraperitoneally. Western blotting, autophagic flux measurement, reactive oxygen species level detection, and coimmunoprecipitation assay were carried out in vitro. Furthermore, to determine the effect of ferritinophagy inhibition on RIA, in vivo knockdown of NCOA4 was carried out by pluronic gel. RESULTS: We verified that accelerated plaque progression was concomitant with endothelial cell (EC) ferroptosis after IR induction, as suggested by a higher level of lipid peroxidation and changes in ferroptosis-associated genes in the PCL + IR group than in the PCL group within vasculature. In vitro experiments further validated the devastating effects of IR on oxidative stress and ferritinophagy in ECs. Mechanistic experiments revealed that IR induced EC ferritinophagy and subsequent ferroptosis in a P38/NCOA4-dependent manner. Both in vitro and in vivo experiments confirmed the therapeutic effect of NCOA4 knockdown in alleviating IR-induced ferritinophagy/ferroptosis of EC and RIA. CONCLUSIONS: Our findings provide novel insights into the regulatory mechanisms of RIA and first prove that IR accelerates atherosclerotic plaque progression by regulating ferritinophagy/ferroptosis of ECs in a P38/NCOA4-dependent manner.


Subject(s)
Ferroptosis , Plaque, Atherosclerotic , Radiation Injuries , Animals , Mice , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Plaque, Atherosclerotic/pathology , Radiotherapy/adverse effects , Radiation Dosage , Radiation Injuries/pathology
6.
Comput Math Methods Med ; 2022: 5382420, 2022.
Article in English | MEDLINE | ID: mdl-36267304

ABSTRACT

Objective: Glomerular endothelium functions as a filtration barrier of metabolites in the kidney. Although X-ray irradiation modulated the permeability of the vascular endothelium, the response of human renal glomerular endothelial cells (HRGECs) to low-dose X-ray irradiation has not been investigated. We evaluated the impacts of low-dose X-ray irradiation on HRGECs and revealed the underlying mechanism. Methods: HRGECs were exposed to X-ray with doses of 0, 0.1, 0.5, 1.0, and 2.0 Gy. The proliferation, viability, and apoptosis of HRGECs were examined by MTT assay, trypan blue staining assay, and TUNEL staining, respectively. The paracellular permeability was assessed by paracellular permeability assay. The expression of VE-cadherin was investigated via immunofluorescence assay. Western blot and qRT-PCR detected the expression levels of VE-cadherin and CLDN5. Besides, the expression levels of pVE-cadherin (pY658), TGF-ß, TGF-ßRI, Src, p-Src, Smad2, p-Smad2, Smad3, p-Smad3, SNAIL, SLUG, and apoptosis-related proteins were tested by Western blot. Results: The proliferation, viability, and apoptosis of HRGECs were not affected by low-dose (<2.0 Gy) X-ray irradiation. X-ray irradiation dose-dependently reduced the level of VE-cadherin, and VE-cadherin and CLDN5 levels were reduced with X-ray irradiation. The levels of pY658, p-Src, p-Smad2, and p-Smad3 were upregulated with the increase in X-ray dose. Besides, the paracellular permeability of HRGECs was increased by even low-dose (<2.0 Gy) X-ray irradiation. Therefore, low-dose X-ray irradiation reduced the cumulative content of VE-cadherin and increased the level of pY658 via activation of the TGF-ß signaling pathway. Conclusion: Even though low-dose X-ray exposure had no impact on proliferation, viability, and apoptosis of HRGECs, it increased the paracellular permeability by deterioration and downregulation of VE-cadherin through stimulating the TGF-ß signaling pathway. This study built the framework for kidney response to low-dose irradiation exposure.


Subject(s)
Endothelial Cells , Trypan Blue , Humans , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , X-Rays , Trypan Blue/metabolism , Cadherins/genetics , Permeability/radiation effects , Kidney , Transforming Growth Factor beta/metabolism
7.
J Photochem Photobiol B ; 235: 112568, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36137302

ABSTRACT

The human corneal endothelial cells (HCEnCs) play a vital role in the maintenance of corneal transparency and visual acuity. In our daily life, HCEnCs are inevitably exposed to ultraviolet B (UVB) radiation leading to decreases of visual acuity and corneal transparency resulting in visual loss eventually. Therefore, understanding the UVB-induced cytotoxicity in HCEnCs is of importance for making efficient strategies to protect our vision from UVB-damage. However, in-depth knowledge about UVB-induced cytotoxicity in HCEnCs is missing. Herein, we pulse-irradiated the HCEnCs in vitro with 150 mJ/cm2 UVB (the environmental dose) at each subculture for 4 passages to explore the insights into UVB-induced phototoxicity. The results showed that the UVB-treated HCEnCs exhibit typical senescent characteristics, including significantly enlarged relative cell area, increased senescence-associated ß-galactosidase positive staining, and upregulated p16INK4A and senescence associated secretory phenotypes (SASPs) such as CCL-27, IL-1α/6/8/10, TGF-ß1 and TNF-α, as well as decreased cell proliferation and Lamin B1 expression, and translocation of Lamin B1. Furthermore, we explored the causative mechanisms of senescence and found that 150 mJ/cm2 UVB pulse-irradiation impairs DNA to activate DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with downregulated DNA repair enzyme PARP1, leading to cell cycle arrest resulting in DDR-mediated senescence. Meanwhile, UVB pulse-irradiation also elicits a consistent increase of ROS production to aggravate DNA damage and impose oxidative stress on energy metabolism leading to metabolic disturbance resulting in metabolic disturbance-mediated senescence. Altogether, the repeated pulse-irradiation of 150 mJ/cm2 UVB induces HCEnC senescence via both DDR pathway and energy metabolism disturbance.


Subject(s)
Cellular Senescence , DNA Damage , Endothelial Cells , Oxidative Stress , Ultraviolet Rays , Cells, Cultured , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/cytology , Endothelial Cells/radiation effects , Humans , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays/adverse effects , beta-Galactosidase/metabolism
8.
Proc Natl Acad Sci U S A ; 119(25): e2202327119, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35696583

ABSTRACT

Pediatric patients with constitutively active mutations in the cytosolic double-stranded-DNA-sensing adaptor STING develop an autoinflammatory syndrome known as STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients have elevated interferon-stimulated gene expression and suffer from interstitial lung disease (ILD) with lymphocyte predominate bronchus-associated lymphoid tissue (BALT). Mice harboring SAVI mutations (STING V154M [VM]) that recapitulate human disease also develop lymphocyte-rich BALT. Ablation of either T or B lymphocytes prolongs the survival of SAVI mice, but lung immune aggregates persist, indicating that T cells and B cells can independently be recruited as BALT. VM T cells produced IFNγ, and IFNγR deficiency prolonged the survival of SAVI mice; however, T-cell-dependent recruitment of infiltrating myeloid cells to the lung was IFNγ independent. Lethally irradiated VM recipients fully reconstituted with wild type bone-marrow-derived cells still developed ILD, pointing to a critical role for VM-expressing radioresistant parenchymal and/or stromal cells in the recruitment and activation of pathogenic lymphocytes. We identified lung endothelial cells as radioresistant cells that express STING. Transcriptional analysis of VM endothelial cells revealed up-regulation of chemokines, proinflammatory cytokines, and genes associated with antigen presentation. Together, our data show that VM-expressing radioresistant cells play a key role in the initiation of lung disease in VM mice and provide insights for the treatment of SAVI patients, with implications for ILD associated with other connective tissue disorders.


Subject(s)
Endothelial Cells , Lung Diseases, Interstitial , Membrane Proteins , T-Lymphocytes , Vascular Diseases , Animals , Child , Endothelial Cells/immunology , Endothelial Cells/radiation effects , Gain of Function Mutation , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/immunology , Lymphocyte Depletion , Lymphoid Tissue/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Radiation Tolerance , T-Lymphocytes/immunology , Vascular Diseases/genetics , Vascular Diseases/immunology
9.
Cell Mol Life Sci ; 79(1): 28, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34936031

ABSTRACT

Microgravity and space radiation (SR) are two highly influential factors affecting humans in space flight (SF). Many health problems reported by astronauts derive from endothelial dysfunction and impaired homeostasis. Here, we describe the adaptive response of human, capillary endothelial cells to SF. Reference samples on the ground and at 1g onboard permitted discrimination between the contribution of microgravity and SR within the combined responses to SF. Cell softening and reduced motility occurred in SF cells, with a loss of actin stress fibers and a broader distribution of microtubules and intermediate filaments within the cytoplasm than in control cells. Furthermore, in space the number of primary cilia per cell increased and DNA repair mechanisms were found to be activated. Transcriptomics revealed the opposing effects of microgravity from SR for specific molecular pathways: SR, unlike microgravity, stimulated pathways for endothelial activation, such as hypoxia and inflammation, DNA repair and apoptosis, inhibiting autophagic flux and promoting an aged-like phenotype. Conversely, microgravity, unlike SR, activated pathways for metabolism and a pro-proliferative phenotype. Therefore, we suggest microgravity and SR should be considered separately to tailor effective countermeasures to protect astronauts' health.


Subject(s)
Autophagy , Capillaries/cytology , Cosmic Radiation , Endothelial Cells/radiation effects , Signal Transduction , Weightlessness , Apoptosis , Biomarkers/metabolism , Cell Line , Cell Survival , Chromosomes, Human/metabolism , Cytoskeleton/metabolism , DNA Damage , Fluorescence , Gene Expression Regulation , Genome, Human , Humans , Male , Mechanotransduction, Cellular , Models, Biological , Signal Transduction/radiation effects , Space Flight , Stress, Physiological , Telomere Homeostasis , Transcriptome/genetics
10.
Cells ; 10(11)2021 11 20.
Article in English | MEDLINE | ID: mdl-34831473

ABSTRACT

Anti-inflammatory low-dose therapy is well established, whereas the immunomodulatory impact of doses below 0.1 Gy is much less clear. In this study, we investigated dose, dose rate and time-dependent effects in a dose range of 0.005 to 2 Gy on immune parameters after whole body irradiation (IR) using a pro-inflammatory (ApoE-/-) and a wild type mouse model. Long-term effects on spleen function (proliferation, monocyte expression) were analyzed 3 months, and short-term effects on immune plasma parameters (IL6, IL10, IL12p70, KC, MCP1, INFγ, TGFß, fibrinogen, sICAM, sVCAM, sE-selectin/CD62) were analyzed 1, 7 and 28 days after Co60 γ-irradiation (IR) at low dose rate (LDR, 0.001 Gy/day) and at high dose rate (HDR). In vitro measurements of murine monocyte (WEHI-274.1) adhesion and cytokine release (KC, MCP1, IL6, TGFß) after low-dose IR (150 kV X-ray unit) of murine endothelial cell (EC) lines (H5V, mlEND1, bEND3) supplement the data. RT-PCR revealed significant reduction of Ki67 and CD68 expression in the spleen of ApoE-/- mice after 0.025 to 2 Gy exposure at HDR, but only after 2 Gy at LDR. Plasma levels in wild type mice, showed non-linear time-dependent induction of proinflammatory cytokines and reduction of TGFß at doses as low as 0.005 Gy at both dose rates, whereas sICAM and fibrinogen levels changed in a dose rate-specific manner. In ApoE-/- mice, levels of sICAM increased and fibrinogen decreased at both dose rates, whereas TGFß increased mainly at HDR. Non-irradiated plasma samples revealed significant age-related enhancement of cytokines and adhesion molecules except for sICAM. In vitro data indicate that endothelial cells may contribute to systemic IR effects and confirm changes of adhesion properties suggested by altered sICAM plasma levels. The differential immunomodulatory effects shown here provide insights in inflammatory changes occurring at doses far below standard anti-inflammatory therapy and are of particular importance after diagnostic and chronic environmental exposures.


Subject(s)
Apolipoproteins E/deficiency , Inflammation/pathology , Radiation, Ionizing , Aging/blood , Animals , Cell Adhesion/radiation effects , Cell Line , Cytokines/metabolism , Dose-Response Relationship, Radiation , Endothelial Cells/radiation effects , Female , Inflammation/blood , Interleukin-6/metabolism , Mice, Inbred C57BL , Monocytes/radiation effects , Spleen/radiation effects , Time Factors
11.
Sci Rep ; 11(1): 18230, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34521931

ABSTRACT

During radiotherapy, microenvironments neighboring the tumor are also exposed to gamma irradiation; this results in unexpected side effects. Blood vessels can serve as microenvironments for tumors and they play an important role in providing nutrients to tumors. This is mostly related to tumor progression, metastasis, and relapse after therapy. Many studies have been performed to obtain a better understanding of tumor vasculature after radiotherapy with in vitro models. However, compared to 3-D models, 2-D in vitro endothelial monolayers cannot physiologically reflect in vivo blood vessels. We previously remodeled the extracellular matrix (ECM) hydrogel that enhanced the tight barrier formation of 3-D blood vessels and the vascular endothelial growth factor (VEGF) gradient induced angiogenesis in a microfluidic device. In this study, the blood vessel model is further introduced to understand how gamma irradiation affects the endothelial monolayer. After the gamma irradiation exposure, we observed a collapsed endothelial barrier and a reduced angiogenic potential. Changes in the cell behaviors of the tip and stalk cells were also detected in the angiogenesis model after irradiation, which is difficult to observe in 2-D monolayer models. Therefore, the 3-D in vitro blood vessel model can be used to understand radiation-induced endothelial injuries.


Subject(s)
Endothelial Cells/radiation effects , Gamma Rays , Neovascularization, Pathologic/metabolism , Tissue Engineering/methods , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Extracellular Matrix/chemistry , Humans , Hydrogels/chemistry , Microfluidics/methods , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
J Histochem Cytochem ; 69(8): 523-534, 2021 08.
Article in English | MEDLINE | ID: mdl-34339312

ABSTRACT

Radiation therapy-mediated salivary gland destruction is characterized by increased inflammatory cell infiltration and fibrosis, both of which ultimately lead to salivary gland hypofunction. However, current treatments (e.g., artificial saliva and sialagogues) only promote temporary relief of symptoms. As such, developing alternative measures against radiation damage is critical for restoring salivary gland structure and function. One promising option for managing radiation therapy-mediated damage in salivary glands is by activation of specialized proresolving lipid mediator receptors due to their demonstrated role in resolution of inflammation and fibrosis in many tissues. Nonetheless, little is known about the presence and function of these receptors in healthy and/or irradiated salivary glands. Therefore, the goal of this study was to detect whether these specialized proresolving lipid mediator receptors are expressed in healthy salivary glands and, if so, if they are maintained after radiation therapy-mediated damage. Our results indicate that specialized proresolving lipid mediator receptors are heterogeneously expressed in inflammatory as well as in acinar and ductal cells within human submandibular glands and that their expression persists after radiation therapy. These findings suggest that epithelial cells as well as resident immune cells represent potential targets for modulation of resolution of inflammation and fibrosis in irradiated salivary glands.


Subject(s)
Radiation Tolerance , Receptors, Chemokine/genetics , Receptors, Formyl Peptide/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Leukotriene B4/genetics , Receptors, Lipoxin/genetics , Submandibular Gland/radiation effects , Acinar Cells/cytology , Acinar Cells/metabolism , Acinar Cells/radiation effects , Adult , Aged , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Female , Gamma Rays , Gene Expression , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Male , Middle Aged , Receptors, Chemokine/metabolism , Receptors, Formyl Peptide/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Leukotriene B4/metabolism , Receptors, Lipoxin/metabolism , Submandibular Gland/cytology , Submandibular Gland/metabolism
13.
Inflamm Res ; 70(10-12): 1141-1150, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34459932

ABSTRACT

UV-B stimulation can induce retinopathy, whose pathogenesis is currently unclear. UV-B mediated inflammation in retinal endothelial cells is reported to be involved in the pathogenesis of retinopathy. S14G-humanin (HNG) is a neuroprotective peptide that has recently been reported to exert significant anti-inflammatory effects and protective properties against cell death. The present study aims to investigate the protective effects of HNG against UV-B-challenged retinal endothelial cells and explore the underlying mechanism. UV-B radiation was used to induce an injury model in human retinal endothelial cells (HRECs). First, exposure to UV-B induced the expression of TXNIP. Additionally, we found that treatment with HNG inhibited the activation of the TXNIP/NLRP3 signaling pathway and mitigated the excessive release of IL-1ß and IL-18 in UV-B-challenged HRECs. UV-B increased the expression of the transcriptional factor endothelial growth response-1 (Egr-1). Interestingly, overexpression of Egr-1 increased the luciferase activity of the TXNIP promoter as well as the mRNA and protein expression of TXNIP. In contrast, the knockdown of Egr-1 reduced the expression of TXNIP under both the normal and UV-B exposure conditions. Importantly, treatment with HNG attenuated UV-B-induced expression of Egr-1. However, overexpression of Egr-1 abolished the inhibitory effects of HNG-induced activation of NLRP3 as well as the production of IL-1ß and IL-18. Taken together, our findings reveal that HNG protected retinal endothelial cells from UV-B-induced NLRP3 inflammation activation through inhibiting TXNIP mediated by Egr-1.


Subject(s)
Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Peptides/pharmacology , Radiation-Protective Agents/pharmacology , Retina/cytology , Ultraviolet Rays , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Endothelial Cells/metabolism , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/genetics , Inflammation/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
14.
Toxicology ; 458: 152841, 2021 06 30.
Article in English | MEDLINE | ID: mdl-34216699

ABSTRACT

The cardiotoxicity of various anticancer therapies, including radiotherapy, can lead to cardiovascular complications. These complications can range from damaging cardiac tissues within the irradiation field to increasing the long-term risks of developing heart failure, coronary artery disease, and myocardial infarction. We analyzed radiation-induced metabolites capable of mediating critical biological processes, such as inflammation, senescence, and apoptosis. Previously, by applying QTOF-MASS analysis to irradiated human fibroblasts, we identified that metabolite sets of lysophosphatidylcholine (LPC) were increased in these cells. In this study, radiation-induced LPC accumulation in human aortic endothelial cells (HAECs) increased reactive oxygen species (ROS) production and senescence-associated-beta-galactosidase staining, in addition to decreasing their tube-forming ability. Knockdown of lipoprotein-associated phospholipase A2 (Lp-PLA2) with small interfering RNA (siRNA) inhibited the increased LPC production induced by radiation, and reduced the radiation-induced cell damage produced by ROS and oxidized low-density lipoprotein (LDL). Lp-PLA2 depletion abolished the induction of proinflammatory factors, such as interleukin 1ß, tumor necrosis factor-alpha, matrix metalloproteinase 2, and matrix metalloproteinase 9, as well as adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) and E-selection. Likewise, we showed that Lp-PLA2 expression was upregulated in the vasculature of irradiated rat, resulting in increased LPC production and LDL oxidation. Our data demonstrate that radiation-induced LPC production is a potential risk factor for cardiotoxicity that is mediated by Lp-PLA2 activity, suggesting that LPC and Lp-PLA2 offer potential diagnostic and therapeutic approaches to cardiovascular damage during radiotherapy.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , 1-Alkyl-2-acetylglycerophosphocholine Esterase/radiation effects , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Lysophosphatidylcholines/metabolism , Phospholipases A2/metabolism , Phospholipases A2/radiation effects , Animals , Aorta/pathology , Aorta/radiation effects , Cytokines/metabolism , Female , Gene Knockdown Techniques , Humans , Inflammation/metabolism , Microtubules/drug effects , Microtubules/radiation effects , RNA, Small Interfering/genetics , RNA, Small Interfering/radiation effects , Radiation, Ionizing , Rats , Rats, Inbred F344 , Reactive Oxygen Species/metabolism
15.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34153091

ABSTRACT

Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E2 (dmPGE2), a longer-acting analogue of PGE2, promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE2 and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE2 and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE2 and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE2, alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin-CD45-CD31+Sca-1- sinusoidal endothelial cells, while combined dmPGE2 and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE2 and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.


Subject(s)
16,16-Dimethylprostaglandin E2/therapeutic use , Acute Radiation Syndrome/drug therapy , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Blood Platelets/drug effects , Endothelial Cells/drug effects , Hemorrhagic Disorders/drug therapy , Lisinopril/therapeutic use , Megakaryocytes/drug effects , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Acute Radiation Syndrome/complications , Animals , Blood Platelets/radiation effects , Bone Marrow/drug effects , Bone Marrow/radiation effects , C-Reactive Protein/analysis , Cesium Radioisotopes , Drug Evaluation, Preclinical , Endothelial Cells/radiation effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/radiation effects , Female , Gamma Rays/adverse effects , Hemorrhagic Disorders/etiology , Megakaryocytes/radiation effects , Mice , Mice, Inbred C57BL , P-Selectin/analysis , Platelet Aggregation/drug effects , Platelet Aggregation/radiation effects , Platelet Factor 4/analysis , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/etiology , Thrombocytopenia/etiology , Thrombopoiesis/radiation effects , Whole-Body Irradiation , von Willebrand Factor/analysis
16.
Nat Commun ; 12(1): 3279, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34078883

ABSTRACT

Targeting the molecular pathways underlying the cardiotoxicity associated with thoracic irradiation and doxorubicin (Dox) could reduce the morbidity and mortality associated with these anticancer treatments. Here, we find that vascular endothelial cells (ECs) with persistent DNA damage induced by irradiation and Dox treatment exhibit a fibrotic phenotype (endothelial-mesenchymal transition, EndMT) correlating with the colocalization of L1CAM and persistent DNA damage foci. We demonstrate that treatment with the anti-L1CAM antibody Ab417 decreases L1CAM overexpression and nuclear translocation and persistent DNA damage foci. We show that in whole-heart-irradiated mice, EC-specific p53 deletion increases vascular fibrosis and the colocalization of L1CAM and DNA damage foci, while Ab417 attenuates these effects. We also demonstrate that Ab417 prevents cardiac dysfunction-related decrease in fractional shortening and prolongs survival after whole-heart irradiation or Dox treatment. We show that cardiomyopathy patient-derived cardiovascular ECs with persistent DNA damage show upregulated L1CAM and EndMT, indicating clinical applicability of Ab417. We conclude that controlling vascular DNA damage by inhibiting nuclear L1CAM translocation might effectively prevent anticancer therapy-associated cardiotoxicity.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cardiomyopathies/prevention & control , Cardiotoxicity/prevention & control , Doxorubicin/toxicity , Gamma Rays/adverse effects , Neural Cell Adhesion Molecule L1/genetics , Animals , Antibiotics, Antineoplastic/toxicity , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiotoxicity/etiology , Cardiotoxicity/genetics , Cardiotoxicity/metabolism , Case-Control Studies , Coculture Techniques , DNA Damage , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Profiling , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , Male , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/radiation effects , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neural Cell Adhesion Molecule L1/metabolism , Signal Transduction , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
17.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802812

ABSTRACT

Targeting tumor vasculature through specific endothelial cell markers represents a promising approach for cancer treatment. Here our aim was to construct an antibiotic resistance gene-free plasmid encoding shRNAs to simultaneously target two endothelial cell markers, CD105 and CD146, and to test its functionality and therapeutic potential in vitro when delivered by gene electrotransfer (GET) and combined with irradiation (IR). Functionality of the plasmid was evaluated by determining the silencing of the targeted genes using qRT-PCR. Antiproliferative and antiangiogenic effects were determined by the cytotoxicity assay tube formation assay and wound healing assay in murine endothelial cells 2H-11. The functionality of the plasmid construct was also evaluated in malignant melanoma tumor cell line B16F10. Additionally, potential activation of immune response was measured by induction of DNA sensor STING and proinflammatory cytokines by qRT-PCR in endothelial cells 2H-11. We demonstrated that the plasmid construction was successful and can efficiently silence the expression of the two targeted genes. As a consequence of silencing, reduced migration rate and angiogenic potential was confirmed in 2H-11 endothelial cells. Furthermore, induction of DNA sensor STING and proinflammatory cytokines were determined, which could add to the therapeutic effectiveness when used in vivo. To conclude, we successfully constructed a novel plasmid DNA with two shRNAs, which holds a great promise for further in vivo testing.


Subject(s)
CD146 Antigen/genetics , Electroporation , Endoglin/genetics , Gene Silencing , Plasmids/genetics , Radiation, Ionizing , Transfection , Animals , Cell Death , Cell Line , Cytokines/metabolism , Endothelial Cells/radiation effects , Membrane Proteins , Mice , Neovascularization, Physiologic/radiation effects
18.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33786610

ABSTRACT

Thoracic radiotherapy is an effective treatment for many types of cancer; however it is also associated with an increased risk of developing cardiovascular disease (CVD), appearing mainly ≥10 years after radiation exposure. The present study investigated acute and early term physiological and molecular changes in the cardiovascular system after ionizing radiation exposure. Female and male ApoE­/­ mice received a single exposure of low or high dose X­ray thoracic irradiation (0.1 and 10 Gy). The level of cholesterol and triglycerides, as well as a large panel of inflammatory markers, were analyzed in serum samples obtained at 24 h and 1 month after irradiation. The secretion of inflammatory markers was further verified in vitro in coronary artery and microvascular endothelial cell lines after exposure to low and high dose of ionizing radiation (0.1 and 5 Gy). Local thoracic irradiation of ApoE­/­ mice increased serum growth differentiation factor­15 (GDF­15) and C­X­C motif chemokine ligand 10 (CXCL10) levels in both female and male mice 24 h after high dose irradiation, which were also secreted from coronary artery and microvascular endothelial cells in vitro. Sex­specific responses were observed for triglyceride and cholesterol levels, and some of the assessed inflammatory markers as detailed below. Male ApoE­/­ mice demonstrated elevated intercellular adhesion molecule­1 and P­selectin at 24 h, and adiponectin and plasminogen activator inhibitor­1 at 1 month after irradiation, while female ApoE­/­ mice exhibited decreased monocyte chemoattractant protein­1 and urokinase­type plasminogen activator receptor at 24 h, and basic fibroblast growth factor 1 month after irradiation. The inflammatory responses were mainly significant following high dose irradiation, but certain markers showed significant changes after low dose exposure. The present study revealed that acute/early inflammatory responses occurred after low and high dose thoracic irradiation. However, further research is required to elucidate early asymptomatic changes in the cardiovascular system post thoracic X­irradiation and to investigate whether GDF­15 and CXCL10 could be considered as potential biomarkers for the early detection of CVD risk in thoracic radiotherapy­treated patients.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/metabolism , Chemokine CXCL10/metabolism , Endothelium, Vascular/radiation effects , Growth Differentiation Factor 15/metabolism , X-Rays , Animals , Apolipoproteins E/deficiency , Atherosclerosis/genetics , Cell Adhesion Molecule-1/genetics , Cell Adhesion Molecule-1/metabolism , Cell Line , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL10/genetics , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/cytology , Female , Fibroblast Growth Factor 1/genetics , Fibroblast Growth Factor 1/metabolism , Growth Differentiation Factor 15/genetics , Humans , Male , Mice , Mice, Inbred C57BL , P-Selectin/genetics , P-Selectin/metabolism , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism
19.
World Neurosurg ; 149: e982-e988, 2021 05.
Article in English | MEDLINE | ID: mdl-33508487

ABSTRACT

BACKGROUND: At present, gamma knife radiosurgery plays an important role in neurosurgical procedures. Gamma knife radiosurgery has been used to treat many types of brain tumors and as a functional intervention. However, gamma knife treatment has a devastating effect on the normal brain parenchyma surrounding the target point. It causes increased vascular permeability, vasodilation, and swelling in endothelial cells. Ozone has antioxidant, antiapoptotic, and anti-inflammatory effects in the body. Thus, we evaluated the radioprotective effects of ozone in rats undergoing gamma knife radiation. METHODS: In the present study, 24 Sprague-Dawley male rats weighing 250-300 g in 3 groups of 8 rats each were used. The rats were selected randomly. The control group did not receive any gamma knife radiation. The other 2 groups received 50 Gy of radiation, with 1 group given ozone treatment and the other group not given ozone treatment after gamma knife radiosurgery. At 12 weeks after gamma knife radiation, the rats were sacrificed with high-dose anesthetic agents and the tissues prepared for evaluation. The slides were evaluated for necrosis, vacuolization, glial proliferation, and vascular proliferation using hematoxylin-eosin staining. Vascular endothelial growth factor (VEGF) and extracellular matrix metalloproteinase inducer (also known as CD147) were evaluated using immunohistochemical staining. RESULTS: VEGF expression in glial tissue was significantly less in the group receiving ozone (χ2 = 15.00; df = 4; P = 0.005) compared with the group that had not received ozone and was similar to the expression in the control group. CONCLUSIONS: The lower expression of VEGF in the group receiving ozone might cause less edema in the surrounding tissue owing to less degradation of vascular permeability in the rat brain tissue.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Capillary Permeability/drug effects , Endothelial Cells/drug effects , Ozone/pharmacology , Radiosurgery/adverse effects , Vasodilation/drug effects , Animals , Basigin/drug effects , Basigin/metabolism , Basigin/radiation effects , Blood-Brain Barrier/radiation effects , Brain/pathology , Brain/radiation effects , Brain Edema , Capillary Permeability/radiation effects , Edema , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Rats , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/radiation effects , Vasodilation/radiation effects
20.
Cells ; 11(1)2021 12 27.
Article in English | MEDLINE | ID: mdl-35011634

ABSTRACT

Anti-inflammatory effects of low-dose irradiation often follow a non-linear dose-effect relationship. These characteristics were also described for the modulation of leukocyte adhesion to endothelial cells. Previous results further revealed a contribution of reactive oxygen species (ROS) and anti-oxidative factors to a reduced leukocyte adhesion. Here, we evaluated the expression of anti-oxidative enzymes and the transcription factor Nrf2 (Nuclear factor-erythroid-2-related factor 2), intracellular ROS content, and leukocyte adhesion in primary human microvascular endothelial cells (HMVEC) upon low-dose irradiation under physiological laminar shear stress or static conditions after irradiation with X-ray or Carbon (C)-ions (0-2 Gy). Laminar conditions contributed to increased mRNA expression of anti-oxidative factors and reduced ROS in HMVEC following a 0.1 Gy X-ray and 0.5 Gy C-ion exposure, corresponding to reduced leukocyte adhesion and expression of adhesion molecules. By contrast, mRNA expression of anti-oxidative markers and adhesion molecules, ROS, and leukocyte adhesion were not altered by irradiation under static conditions. In conclusion, irradiation of endothelial cells with low doses under physiological laminar conditions modulates the mRNA expression of key factors of the anti-oxidative system, the intracellular ROS contents of which contribute at least in part to leucocyte adhesion, dependent on the radiation source.


Subject(s)
Endothelial Cells/cytology , Leukocytes/cytology , Microvessels/cytology , Reactive Oxygen Species/metabolism , Carbon , Cell Adhesion/radiation effects , Cell Adhesion Molecules/metabolism , Cells, Cultured , Dose-Response Relationship, Radiation , Endothelial Cells/radiation effects , Gene Expression Regulation/radiation effects , Humans , Leukocytes/radiation effects , Models, Biological , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Oxidative Stress/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , X-Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...