Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 500
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Nucleic Acids Res ; 47(14): 7294-7305, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31251810

ABSTRACT

Cellular senescence, an integral component of aging and cancer, arises in response to diverse triggers, including telomere attrition, macromolecular damage and signaling from activated oncogenes. At present, senescent cells are identified by the combined presence of multiple traits, such as senescence-associated protein expression and secretion, DNA damage and ß-galactosidase activity; unfortunately, these traits are neither exclusively nor universally present in senescent cells. To identify robust shared markers of senescence, we have performed RNA-sequencing analysis across eight diverse models of senescence triggered in human diploid fibroblasts (WI-38, IMR-90) and endothelial cells (HUVEC, HAEC) by replicative exhaustion, exposure to ionizing radiation or doxorubicin, and expression of the oncogene HRASG12V. The intersection of the altered transcriptomes revealed 50 RNAs consistently elevated and 18 RNAs consistently reduced across all senescence models, including many protein-coding mRNAs and some non-coding RNAs. We propose that these shared transcriptome profiles will enable the identification of senescent cells in vivo, the investigation of their roles in aging and malignancy and the development of strategies to target senescent cells therapeutically.


Subject(s)
Cellular Senescence/genetics , Endothelial Cells/metabolism , Fibroblasts/metabolism , Transcriptome , Aging/genetics , Antibiotics, Antineoplastic/pharmacology , Cell Line , Cells, Cultured , Doxorubicin/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Fibroblasts/drug effects , Fibroblasts/radiation effects , Humans , Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation, Ionizing , Sequence Analysis, RNA/methods
6.
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
7.
Cell Mol Life Sci ; 76(4): 699-728, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30377700

ABSTRACT

The endothelium, a tissue that forms a single layer of cells lining various organs and cavities of the body, especially the heart and blood as well as lymphatic vessels, plays a complex role in vascular biology. It contributes to key aspects of vascular homeostasis and is also involved in pathophysiological processes, such as thrombosis, inflammation, and hypertension. Epidemiological data show that high doses of ionizing radiation lead to cardiovascular disease over time. The aim of this review is to summarize the current knowledge on endothelial cell activation and dysfunction after ionizing radiation exposure as a central feature preceding the development of cardiovascular diseases.


Subject(s)
Endothelial Cells/radiation effects , Endothelium, Vascular/radiation effects , Endothelium/radiation effects , Radiation Injuries/physiopathology , Radiation, Ionizing , Animals , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cellular Senescence/radiation effects , Endothelial Cells/pathology , Endothelium/pathology , Endothelium/physiopathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Models, Biological
8.
Int J Mol Sci ; 21(7)2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32230970

ABSTRACT

Normal tissue toxicity is a dose-limiting factor in radiation therapy. Therefore, a detailed understanding of the normal tissue response to radiation is necessary to predict the risk of normal tissue toxicity and to development strategies for tissue protection. One component of normal tissue that is continuously exposed during therapeutic irradiation is the circulating population of peripheral blood mononuclear cells (PBMC). PBMCs are highly sensitive to ionizing radiation (IR); however, little is known about how IR affects the PBMC response on a systemic level. It was the aim of this study to investigate whether IR was capable to induce changes in the composition and function of extracellular vesicles (EVs) secreted from PBMCs after radiation exposure to different doses. Therefore, whole blood samples from healthy donors were exposed to X-ray radiation in the clinically relevant doses of 0, 0.1, 2 or 6 Gy and PBMC-secreted EVs were isolated 72 h later. Proteome and miRNome analysis of EVs as well as functional studies were performed. Secreted EVs showed a dose-dependent increase in the number of significantly deregulated proteins and microRNAs. For both, proteome and microRNA data, principal component analysis showed a dose-dependent separation of control and exposed groups. Integrated pathway analysis of the radiation-regulated EV proteins and microRNAs consistently predicted an association of deregulated molecules with apoptosis, cell death and survival. Functional studies identified endothelial cells as an efficient EV recipient system, in which irradiation of recipient cells further increased the uptake. Furthermore an apoptosis suppressive effect of EVs from irradiated PBMCs in endothelial recipient cells was detected. In summary, this study demonstrates that IR modifies the communication between PBMCs and endothelial cells. EVs from irradiated PBMC donors were identified as transmitters of protective signals to irradiated endothelial cells. Thus, these data may lead to the discovery of biomarker candidates for radiation dosimetry and even more importantly, they suggest EVs as a novel systemic communication pathway between irradiated normal, non-cancer tissues.


Subject(s)
Extracellular Vesicles/metabolism , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/radiation effects , Radiation Exposure , Secretory Vesicles/metabolism , Apoptosis/radiation effects , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Humans , MicroRNAs/genetics , Proteome/metabolism , Radiation, Ionizing , Radiotherapy/methods
9.
Int J Mol Sci ; 21(5)2020 Mar 10.
Article in English | MEDLINE | ID: mdl-32164317

ABSTRACT

Although radiotherapy plays a crucial in the management of pelvic tumors, its toxicity on surrounding healthy tissues such as the small intestine, colon, and rectum is one of the major limitations associated with its use. In particular, proctitis is a major clinical complication of pelvic radiotherapy. Recent evidence suggests that endothelial injury significantly affects the initiation of radiation-induced inflammation. The damaged endothelial cells accelerate immune cell recruitment by activating the expression of endothelial adhesive molecules, which participate in the development of tissue damage. Pravastatin, a cholesterol lowering drug, exerts persistent anti-inflammatory and anti-thrombotic effects on irradiated endothelial cells and inhibits the interaction of leukocytes and damaged endothelial cells. Here, we aimed to investigate the effects of pravastatin on radiation-induced endothelial damage in human umbilical vein endothelial cell and a murine proctitis model. Pravastatin attenuated epithelial damage and inflammatory response in irradiated colorectal lesions. In particular, pravastatin improved radiation-induced endothelial damage by regulating thrombomodulin (TM) expression. In addition, exogenous TM inhibited leukocyte adhesion to the irradiated endothelial cells. Thus, pravastatin can inhibit endothelial damage by inducing TM, thereby alleviating radiation proctitis. Therefore, we suggest that pharmacological modulation of endothelial TM may limit intestinal inflammation after irradiation.


Subject(s)
Endothelial Cells/cytology , Pravastatin/administration & dosage , Proctitis/drug therapy , Thrombomodulin/metabolism , Animals , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , Female , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Mice , Pravastatin/pharmacology , Proctitis/etiology , THP-1 Cells
10.
BMC Cancer ; 19(1): 958, 2019 Oct 16.
Article in English | MEDLINE | ID: mdl-31619190

ABSTRACT

BACKGROUND: We analyzed the changes in permeability of endothelial cell layers after photon irradiation, with a focus on the metalloproteases ADAM10 and ADAM17, and on VE-cadherin, components crucial for the integrity of endothelial intercellular junctions, and their roles in the transmigration of cancer cells through endothelial cell monolayers. METHODS: Primary HUVEC were irradiated with 2 or 4 Gy photons at a dose rate of 5 Gy/min. The permeability of an irradiated endothelial monolayer for macromolecules and tumor cells was analyzed in the presence or absence of the ADAM10/17 inhibitors GI254023X and GW280264X. Expression of ADAM10, ADAM17 and VE-Cadherin in endothelial cells was quantified by immunoblotting and qRT. VE-Cadherin was additionally analyzed by immunofluorescence microscopy and ELISA. RESULTS: Ionizing radiation increased the permeability of endothelial monolayers and the transendothelial migration of tumor cells. This was effectively blocked by a selective inhibition (GI254023X) of ADAM10. Irradiation increased both, the expression and activity of ADAM10, which led to increased degradation of VE-cadherin, but also led to higher rates of VE-cadherin internalization. Increased degradation of VE-cadherin was also observed when endothelial monolayers were exposed to tumor-cell conditioned medium, similar to when exposed to recombinant VEGF. CONCLUSIONS: Our results suggest a mechanism of irradiation-induced increased permeability and transendothelial migration of tumor cells based on the activation of ADAM10 and the subsequent change of endothelial permeability through the degradation and internalization of VE-cadherin.


Subject(s)
ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/radiation effects , Human Umbilical Vein Endothelial Cells/radiation effects , Membrane Proteins/metabolism , Proteolysis/radiation effects , Radiation, Ionizing , Transendothelial and Transepithelial Migration/radiation effects , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/genetics , ADAM17 Protein/antagonists & inhibitors , ADAM17 Protein/genetics , ADAM17 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/genetics , Cell Line, Tumor , Dipeptides/pharmacology , Endothelial Cells/metabolism , Humans , Hydroxamic Acids/pharmacology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Permeability/radiation effects , Radiotherapy/adverse effects , Signal Transduction/radiation effects , Transendothelial and Transepithelial Migration/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology
11.
Biol Res ; 52(1): 22, 2019 Apr 16.
Article in English | MEDLINE | ID: mdl-30992075

ABSTRACT

BACKGROUND: Tumor microenvironment (TME) plays a vital role in determining the outcomes of radiotherapy. As an important component of TME, vascular endothelial cells are involved in the perivascular resistance niche (PVRN), which is formed by inflammation or cytokine production induced by ionizing radiation (IR). Protein kinase CK2 is a constitutively active serine/threonine kinase which plays a vital role in cell proliferation and inflammation. In this study, we investigated the potential role of CK2 in PVRN after IR exposure. RESULT: Specific CK2 inhibitors, Quinalizarin and CX-4945, were employed to effectively suppressed the kinase activity of CK2 in human umbilical vein endothelial cells (HUVECs) without affecting their viability. Results showing that conditioned medium from IR-exposed HUVECs increased cell viability of A549 and H460 cells, and the pretreatment of CK2 inhibitors slowed down such increment. The secretion of IL-8 and IL-6 in HUVECs was induced after exposure with IR, but significantly inhibited by the addition of CK2 inhibitors. Furthermore, IR exposure elevated the nuclear phosphorylated factor-κB (NF-κB) p65 expression in HUVECs, which was a master factor regulating cytokine production. But when pretreated with CK2 inhibitors, such elevation was significantly suppressed. CONCLUSION: This study indicated that protein kinase CK2 is involved in the key process of the IR induced perivascular resistant niche, namely cytokine production, by endothelial cells, which finally led to radioresistance of non-small cell lung cancer cells. Thus, the inhibition of CK2 may be a promising way to improve the outcomes of radiation in non-small cell lung cancer cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Casein Kinase II/antagonists & inhibitors , Endothelial Cells/radiation effects , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Anthraquinones/pharmacology , Blotting, Western , Cytokines/biosynthesis , Endothelium, Vascular/cytology , Humans , Naphthyridines/pharmacology , Phenazines
12.
Lasers Med Sci ; 34(3): 495-504, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30145725

ABSTRACT

Photobiomodulation of cells using near-infrared (NIR) monochromatic light can affect cell functions such as proliferation, viability, and metabolism in a range of cell types. Evidence for the effects of near-infrared light on endothelial cells has been reported, but the studies were mainly performed using VIS light emitted by low-energy lasers, because NIR wavelengths seemed negatively stimulate these cells. Cell viability, free radical-induced oxidative stress, NF-κB activation, nitric oxide release, mitochondrial respiration, and wound healing repair were assessed in human endothelial cells (HECV) irradiated with 808-nm diode laser light (laser setup = 1 W/cm2, 60 s, 60 J/cm2, CW vs measured energy parameter = 0.95 W/cm2, 60 s, 57 J/cm2, mode CW) emitted by an handpiece with flat-top profile. No difference in viability was detected between controls and HECV cells irradiated with 808-nm diode laser light for 60 s. Irradiated cells demonstrated higher proliferation rate and increased migration ability associated to moderate increase in ROS production without a significant increase in oxidative stress and oxidative stress-activated processes. Near-infrared light stimulated mitochondrial oxygen consumption and ATP synthesis in HECV cells. Short near-infrared irradiation did not affect viability of HECV cells, rather led to a stimulation of wound healing rate, likely sustained by ROS-mediated stimulation of mitochondrial activity. Our results demonstrating that near-infrared led to a shift from anaerobic to aerobic metabolism provide new insight into the possible molecular mechanisms by which photobiomodulation with 808-nm diode laser light protects against inflammation-induced endothelial dysfunction, seemingly promising to enhance their therapeutic properties.


Subject(s)
Endothelial Cells/radiation effects , Lasers, Semiconductor , Low-Level Light Therapy , Mitochondria/metabolism , Mitochondria/radiation effects , Oxidative Phosphorylation/radiation effects , Reactive Oxygen Species/metabolism , Wound Healing/radiation effects , Aerobiosis , Cell Line , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Endothelial Cells/metabolism , Humans , Nitric Oxide/metabolism
13.
Int J Mol Sci ; 20(23)2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31757032

ABSTRACT

Vascular targeting with pro-thrombotic antibody-conjugates is a promising biological treatment for brain arteriovenous malformations (bAVMs). However, targeted drug delivery relies on the identification of unique or overexpressed markers on the surface of a target cell. In the absence of inherent biological markers, stereotactic radiosurgery may be used to prime induction of site-specific and targetable molecular changes on the endothelial surface. To investigate lumen-accessible, endothelial targets induced by radiation, we combined Gamma knife surgery in an AVM animal model with in vivo biotin-labeling and comparative proteomics. Two proteins, αB-crystallin (CRYAB)-a small heat shock protein that normally acts as an intracellular chaperone to misfolded proteins-and activated leukocyte cell adhesion molecule CD166, were further validated for endothelial surface expression after irradiation. Immunostaining of endothelial cells in vitro and rat AVM tissue ex vivo confirmed de novo induction of CRYAB following irradiation (20 Gy). Western analysis demonstrated that CRYAB accumulated intracellularly as a 20 kDa monomer, but, at the cell surface, a novel 65 kDa protein was observed, suggesting radiation stimulates translocation of an atypical CRYAB isoform. In contrast, CD166 had relatively high expression in non-irradiated cells, localized predominantly to the lateral surfaces. Radiation increased CD166 surface exposure by inducing translocation from intercellular junctions to the apical surface without significantly altering total protein levels. These findings reinforce the dynamic molecular changes induced by radiation exposure, particularly at the cell surface, and support further investigation of radiation as a priming mechanism and these molecules as putative targets for focused drug delivery in irradiated tissue.


Subject(s)
Crystallins/metabolism , Endothelial Cells/radiation effects , Intracranial Arteriovenous Malformations/radiotherapy , Microtubule-Associated Proteins/metabolism , Radiosurgery/adverse effects , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Cell Membrane/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Gamma Rays/adverse effects , Intracranial Arteriovenous Malformations/metabolism , Mice , Protein Transport , Rats , Rats, Sprague-Dawley
14.
Int J Mol Sci ; 20(7)2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30978983

ABSTRACT

The bone marrow (BM) microenvironment plays a crucial role in the maintenance and regeneration of hematopoietic stem (HSC) and progenitor cells (HSPC). In particular, the vascular niche is responsible for regulating HSC maintenance, differentiation, and migration of cells in and out of the BM. Damage to this niche upon exposure to ionizing radiation, whether accidental or as a result of therapy, can contribute to delays in HSC recovery and/or function. The ability of BM derived-endothelial cells (BMEC) to alter and/or protect HSPC after exposure to ionizing radiation was investigated. Our data show that exposure of BMEC to ionizing radiation resulted in alterations in Akt signaling, increased expression of PARP-1, IL6, and MCP-1, and decreased expression of MMP1 and MMP9. In addition, global analysis of gene expression of HSC and BMEC in response to mixed neutron/gamma field (MF) radiation identified 60 genes whose expression was altered after radiation in both cell types, suggesting that a subset of genes is commonly affected by this type of radiation. Focused gene analysis by RT-PCR revealed two categories of BMEC alterations: (a) a subset of genes whose expression was altered in response to radiation, with no additional effect observed during coculture with HSPC, and (b) a subset of genes upregulated in response to radiation, and altered when cocultured with HSPC. Coculture of BMEC with CD34+ HSPC induced HSPC proliferation, and improved BM function after MF radiation. Nonirradiated HSPC exhibited reduced CD34 expression over time, but when irradiated, they maintained higher CD34 expression. Nonirradiated HSPC cocultured with nonirradiated BMEC expressed lower levels of CD34 expression compared to nonirradiated alone. These data characterize the role of each cell type in response to MF radiation and demonstrate the interdependence of each cell's response to ionizing radiation. The identified genes modulated by radiation and coculture provide guidance for future experiments to test hypotheses concerning specific factors mediating the beneficial effects of BMEC on HSPC. This information will prove useful in the search for medical countermeasures to radiation-induced hematopoietic injury.


Subject(s)
Bone Marrow Cells/radiation effects , Coculture Techniques , Endothelial Cells/radiation effects , Hematopoietic Stem Cells/radiation effects , Antigens, CD34/analysis , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/radiation effects , Cell Line , Cell Proliferation/radiation effects , Coculture Techniques/methods , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gamma Rays , Gene Expression Regulation/radiation effects , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Neutrons , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Radiation Injuries/prevention & control , Signal Transduction/radiation effects
15.
Mod Pathol ; 31(9): 1361-1366, 2018 09.
Article in English | MEDLINE | ID: mdl-29713042

ABSTRACT

We hypothesized that in glioblastoma recurring after radiotherapy, a condition whereby the brain endothelium undergoes radiation-induced senescence, tumor cells with endothelial phenotype may be relevant for tumor neovascularization. Matched glioblastoma samples obtained at primary surgery and at surgery for tumor recurrence after radiotherapy, all expressing epidermal growth factor receptor variant III (EGFRvIII), were assessed by a technique that combines fluorescent in situ hybridization (FISH) for the EGFR/CEP7 chromosomal probe with immunostaining for endothelial cells (CD31) and activated pericytes (α Smooth Muscle Actin). Five EGFRvIII-expressing paired primary/recurrent glioblastoma samples, in which the tumor cells showed EGFR/CEP7 amplification, were then assessed by CD31 and α Smooth Muscle Actin immunofluorescence. In glomeruloid bodies, the ratio between CD31+ cells with amplified EGFR/CEP7 signal and the total CD31+ cells was 0.23 ± 0.09 (mean ± sem) and 0.63 ± 0.07 in primary tumors and in recurrent ones, respectively (p < 0.002, Student-t test). In capillaries, the ratio of CD31+ cells with amplified EGFR/CEP7 over the total CD31+ cells lining the capillary lumen was 0.21 ± 0.06 (mean ± sem) and 0.42 ± 0.07 at primary surgery and at recurrence, respectively (p < 0.005, Student-t test). Expression of α Smooth Muscle Actin by cells with EGFR/CEP7 amplification was not observed. Then, in glioblastoma recurring after radiotherapy, where the brain endothelium suffers from radiation-induced cell senescence, tumor-derived endothelium plays a role in neo-vascularization.


Subject(s)
Brain Neoplasms/pathology , Cell Transdifferentiation/physiology , Endothelial Cells/pathology , Glioblastoma/pathology , Neoplasm Recurrence, Local/pathology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/radiotherapy , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , In Situ Hybridization, Fluorescence , Mice , Neoplasm Recurrence, Local/genetics
16.
Exp Dermatol ; 27(9): 973-980, 2018 09.
Article in English | MEDLINE | ID: mdl-29742305

ABSTRACT

Red grape (Vitis vinifera L.) flavonoids including flavan-3-ols (eg, catechin and epicatechin), flavonols (eg, quercetin) and anthocyanins (eg, malvidin) exert anti-inflammatory and antioxidant activities. In the skin they also have a photoprotective action, and their effects have been extensively investigated in keratinocytes, melanocytes and fibroblasts. Despite their known effects also on blood vasculature, little is known on their activities on human dermal blood endothelial cells (HDBECs), which are critically involved in skin homeostasis as well as in the pathogenesis of neoplastic and inflammatory skin diseases. We sought to study the biological effects of selected red grape flavonoids in preventing the consequences of ultraviolet (UV)-A irradiation in vitro. Our results show that red grape flavonoids prevent UV-A-induced sICAM-1 release in HDBECs, suggesting that this cell type could represent an additional target of the anti-inflammatory activity of flavonoids. In addition, flavonoids effectively inhibited UV-A-induced synthesis of collagen type III at both RNA and protein level, indicating that dermal blood microvasculature could be actively involved in ECM remodelling as a consequence of skin photo-ageing, and that this can be prevented by red grape flavonoids.


Subject(s)
Collagen Type III/biosynthesis , Endothelial Cells/drug effects , Endothelial Cells/radiation effects , Flavonoids/pharmacology , Plant Extracts/pharmacology , Vitis , Catechin/pharmacology , Cell Survival/drug effects , Cells, Cultured , Collagen Type III/genetics , Fruit , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Intercellular Adhesion Molecule-1/metabolism , Pyrimidine Dimers/metabolism , Quercetin/analogs & derivatives , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Skin/cytology , Ultraviolet Rays , Up-Regulation/drug effects , Up-Regulation/radiation effects
17.
Lasers Med Sci ; 33(2): 279-286, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29098460

ABSTRACT

Low-level laser therapy (LLLT), widely used in physiotherapy, has been known to enhance wound healing and stimulate cell proliferation, including fibroblast and endothelial cells. Applying LLLT can increase cell proliferation in many kinds of cells including fibroblasts and endothelial cells. However, the protective mechanisms of LLLT on endothelial apoptosis remain unclear. We hypothesized LLLT can protect endothelial cells from inflammation-induced apoptosis. Human endothelial cell line, EA.hy926 cells, and TNF-α/cycloheximide (TNF/CHX) were used to explore the protective effects of LLLT (660 nm) on inflammation-induced endothelial apoptosis. Cell viability, apoptosis, caspase-3/7/8/9 activity, MAPKs signaling, NF-κB activity, and inducible/endothelial nitric oxide synthase (iNOS/eNOS) expression were measured. Our results showed that LLLT increased EA.hy926 cell proliferation, attenuated the TNF/CHX-induced apoptosis, and reduced the TNF/CHX-mediated caspase-3/7/8/9 activation. In addition, LLLT increased ERK MAPK phosphorylation and suppressed the TNF/CHX-increased p38 MAPK, JNK, IKK phosphorylation, NF-κB translocation, and iNOS expression. The caspases-3 cleavage and cell death were not increased in cells treating with ERK inhibitor U0126, which implicated that ERK is not to be responsible for the protective effects of LLLT. After treating with p38 mitogen-activated protein kinase (MAPK) activator, the protection of LLLT in cell apoptosis was no longer existed, showing that LLLT protected the endothelial cells by suppressing p38 MAPK signaling. Our results provide a new insight into the possible molecular mechanisms in which LLLT protects against inflammatory-induced endothelial dysfunction.


Subject(s)
Apoptosis/radiation effects , Cycloheximide/adverse effects , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Low-Level Light Therapy , Tumor Necrosis Factor-alpha/adverse effects , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Endothelial Cells/drug effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , p38 Mitogen-Activated Protein Kinases/metabolism
18.
Clin Oral Investig ; 22(6): 2251-2262, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29307045

ABSTRACT

OBJECTIVES: Head and neck squamous cell carcinoma (HNSCC) shows increased radioresistance due to the manipulation of homeostatic mechanisms like the heat shock response. This study intended to comparatively analyze effects of ionizing radiation on different HNSCC cell lines (PCI) and normal human dermal fibroblasts (NHFs) and human dermal microvascular endothelial cells (HDMECs) to uncover differences in radiation coping strategies. MATERIALS AND METHODS: Proliferation (BrdU assay), apoptosis (caspase 3/7) and intracellular protein expression of heat shock protein (HSP)-70, and phosphorylated and total HSP27, determined by enzyme-linked immunosorbent assay (ELISA), were analyzed after exposure to increasing doses of ionizing radiation (2, 6, and 12 Gray, Gy). RESULTS: Cell count decreased dose-dependently, but PCI cell lines consistently showed higher numbers compared to NHF and HDMEC. Likewise, high doses reduced cell proliferation, but low-dose radiation (2 Gy) instead increased proliferation in PCI 9 and 52. Apoptosis was not detectable in PCI cell lines. Basic HSP70 expression was high in PCI cells with little additional increase by irradiation. PCI cells yielded high basic total HSP27 concentrations but irradiation dose-dependently increased HSP27 in HDMEC, NHF, and PCI cells. Phosphorylated HSP27 concentrations were highest in NHF. CONCLUSION: PCI cell lines showed higher resistance to dose-dependent reduction in cell number, proliferation, and protection from apoptosis compared to NHF and HDMEC. In parallel, we observed a high basic and radiation-induced expression of intracellular HSP70 leading to the assumption that the radioresistance of PCI cells is conferred by HSP70. CLINICAL RELEVANCE: HNSCC use HSP to escape radiation-induced apoptosis and certain subtypes might increase proliferation after low-dose irradiation.


Subject(s)
Apoptosis/radiation effects , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , Endothelial Cells/radiation effects , Fibroblasts/radiation effects , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Heat-Shock Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Enzyme-Linked Immunosorbent Assay , Humans
19.
J Proteome Res ; 16(10): 3903-3916, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28849662

ABSTRACT

Radiation is the most common treatment of cancer. Minimizing the normal tissue injury, especially the damage to vascular endothelium, remains a challenge. This study aimed to analyze direct and indirect radiation effects on the endothelium by investigating mechanisms of signal transfer from irradiated to nonirradiated endothelial cells by means of secreted proteins. Human coronary artery endothelial cells (HCECest2) undergo radiation-induced senescence in vitro 14 days after exposure to 10 Gy X-rays. Proteomics analysis was performed on HCECest2 14 days after irradiation with X-ray doses of 0 Gy (control) or 10 Gy using label-free technology. Additionally, the proteomes of control and radiation-induced secretomes, and those of nonirradiated HCECest2 exposed for 24 h to secreted proteins of either condition were measured. Key changes identified by proteomics and bioinformatics were validated by immunoblotting, ELISA, bead-based multiplex assays, and targeted transcriptomics. The irradiated cells, their secretome, and the nonirradiated recipient cells showed similar inflammatory response, characterized by induction of interferon type I-related proteins and activation of the STAT3 pathway. These data indicate that irradiated endothelial cells may adversely affect nonirradiated surrounding cells via senescence-associated secretory phenotype. This study adds to our knowledge of the pathological background of radiation-induced cardiovascular disease.


Subject(s)
Inflammation/genetics , Neoplasms/radiotherapy , Proteome/genetics , Radiotherapy/adverse effects , STAT3 Transcription Factor/genetics , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Coronary Vessels/metabolism , Coronary Vessels/pathology , Coronary Vessels/radiation effects , Dose-Response Relationship, Radiation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Inflammation/etiology , Inflammation/pathology , Male , Neoplasms/complications , Neoplasms/genetics , Proteome/radiation effects , Proteomics/methods , Signal Transduction/radiation effects
20.
Environ Res ; 156: 297-305, 2017 07.
Article in English | MEDLINE | ID: mdl-28376375

ABSTRACT

PURPOSE: The clonogenic property and radiobiological responses of a fish brain endothelial cell line, eelB, derived from the American eel were studied. METHODS: Clonogenic assays were performed to determine the plating efficiency of the eelB cells and to evaluate the clonogenic survival fractions after direct irradiation to low-dose low-LET gamma radiation or receiving irradiated cell conditioned medium in the bystander effect experiments. RESULT: eelB had the second highest plating efficiency ever reported to date for fish cell lines. Large eelB macroscopic colonies could be formed in a short period of time and were easy to identify and count. Unlike with other fish clonogenic cell lines, which had a relatively slow proliferation profile, clonogenic assays with the eelB cells could be completed as early as 12 days in culture. After direct irradiation with gamma rays at low doses ranging from 0.1Gy to 5Gy, the dose-clonogenic survival curve of the eelB cell line showed a linear trend and did not develop a shoulder region. A classical radio-adaptive response was not induced with the clonogenic survival endpoint when the priming dose (0.1 or 0.5Gy) was delivered 6h before the challenge dose (3 or 5Gy). However, a radio-adaptive response was observed in progeny cells that survived 5Gy and developed lethal mutations. eelB appeared to lack the ability to produce damaging radiation-induced bystander signals on both eelB and HaCaT recipient cells. CONCLUSION: eelB cell line could be a very useful cell model in the study of radiation impacts on the aquatic health.


Subject(s)
Anguilla , Brain/radiation effects , Endothelial Cells/radiation effects , Gamma Rays/adverse effects , Animals , Bystander Effect , Cell Line , Dose-Response Relationship, Radiation , Genomic Instability/radiation effects , Mutation/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL