Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
PLoS One ; 19(6): e0303434, 2024.
Article in English | MEDLINE | ID: mdl-38865377

ABSTRACT

The modulation of macrophage polarization is a promising strategy for maintaining homeostasis and improving innate and adaptive immunity. Low-dose ionizing radiation has been implicated in macrophage immunomodulatory responses. However, studies on the relationship between exosomes and regulation of macrophage polarization induced by ionizing radiation are limited. Therefore, this study investigated the alterations in macrophages and exosomes induced by gamma irradiation and elucidated the underlying mechanisms. We used the mouse macrophage cell line RAW 264.7 to generate macrophages and performed western blot, quantitative reverse transcription-PCR, and gene ontology analyses to elucidate the molecular profiles of macrophage-derived exosomes under varying treatment conditions, including 10 Gy gamma irradiation. Exosomes isolated from gamma-irradiated M1 macrophages exhibited an enhanced M1 phenotype. Irradiation induced the activation of NF-κB and NLRP3 signaling in M1 macrophages, thereby promoting the expression of pro-inflammatory cytokines. Cytokine expression was also upregulated in gamma-irradiated M1 macrophage-released exosomes. Therefore, gamma irradiation has a remarkable effect on the immunomodulatory mechanisms and cytokine profiles of gamma-irradiated M1 macrophage-derived exosomes, and represents a potential immunotherapeutic modality.


Subject(s)
Cytokines , Exosomes , Gamma Rays , Macrophages , Animals , Exosomes/metabolism , Exosomes/radiation effects , Mice , Macrophages/radiation effects , Macrophages/immunology , Macrophages/metabolism , RAW 264.7 Cells , Cytokines/metabolism , NF-kappa B/metabolism , Signal Transduction/radiation effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Macrophage Activation/radiation effects
2.
BMC Cancer ; 24(1): 664, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822331

ABSTRACT

Recent studies have shown that blue light-emitting diode (LED) light has anti-tumor effects, suggesting the possibility of using visible light in cancer therapy. However, the effects of blue light irradiation on cells in the tumor microenvironment, including tumor-associated macrophages (TAMs), are unknown. Here, THP-1 cells were cultured in the conditioned medium (CM) of HCT-116 cells to prepare TAMs. TAMs were divided into LED-irradiated and control groups. Then, the effects of blue LED irradiation on TAM activation were examined. Expression levels of M2 macrophage markers CD163 and CD206 expression were significantly decreased in LED-irradiated TAMs compared with the control group. While control TAM-CM could induce HCT-116 cell migration, these effects were not observed in cells cultured in TAM-CM with LED irradiation. Vascular endothelial growth factor (VEGF) secretion was significantly suppressed in LED-exposed TAMs. PD-L1 expression was upregulated in HCT-116 cells cultured with TAM-CM but attenuated in cells cultured with LED-irradiated TAM-CM. In an in vivo model, protein expression levels of F4/80 and CD163, which are TAM markers, were reduced in the LED-exposed group. These results indicate that blue LED light may have an inhibitory effect on TAMs, as well as anti-tumor effects on colon cancer cells.


Subject(s)
Blue Light , Colonic Neoplasms , Tumor-Associated Macrophages , Animals , Humans , Mice , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Cell Movement/radiation effects , Colonic Neoplasms/radiotherapy , Culture Media, Conditioned/pharmacology , HCT116 Cells , Macrophage Activation/radiation effects , Macrophages/metabolism , Macrophages/radiation effects , Phototherapy/methods , Receptors, Cell Surface/metabolism , Tumor Microenvironment/radiation effects , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/radiation effects , Vascular Endothelial Growth Factor A/metabolism
3.
Adv Sci (Weinh) ; 10(5): e2205048, 2023 02.
Article in English | MEDLINE | ID: mdl-36515274

ABSTRACT

Intelligent control of the immune response is essential for obtaining percutaneous implants with good sterilization and tissue repair abilities. In this study, polypyrrole (Ppy) nanoparticles enveloping a 3D frame of sulfonated polyether ether ketone (SP) surface are constructed, which enhance the surface modulus and hardness of the sulfonated layer by forming a cooperative structure of simulated reinforced concrete and exhibit a superior photothermal effect. Ppy-coated SP could quickly accumulate heat on the surface by responding to 808 nm near-infrared (NIR) light, thereby killing bacteria, and destroying biofilms. Under NIR stimulation, the phagocytosis and M1 activation of macrophages cultured on Ppy-coated SP are enhanced by activating complement 3 and its receptor, CD11b. Phagocytosis and M1 activation are impaired along with abolishment of NIR stimulation in the Ppy-coated SP group, which is favorable for tissue repair. Ppy-coated SP promotes Collagen-I, vascular endothelial growth factor, connective tissue growth factor, and α-actin (Acta2) expression by inducing M2 polarization owing to its higher surface modulus. Overall, Ppy-coated SP with enhanced mechanical properties could be a good candidate for clinical percutaneous implants through on-off phagocytosis and switchable macrophage activation stimulated with NIR.


Subject(s)
Infrared Rays , Macrophage Activation , Nanoparticles , Phagocytosis , Polymers , Pyrroles , Ketones , Macrophage Activation/radiation effects , Phagocytosis/radiation effects , Polyethylene Glycols , Polymers/chemistry , Pyrroles/chemistry , Vascular Endothelial Growth Factor A , Infrared Rays/therapeutic use , Nanoparticles/therapeutic use , Mice , Animals
4.
J Invest Dermatol ; 141(8): 2056-2066.e10, 2021 08.
Article in English | MEDLINE | ID: mdl-33676947

ABSTRACT

Near-infrared (NIR) can penetrate the dermis. NIR is able to regulate cutaneous component cells and immune cells and shows significant anti-inflammatory therapeutic effects. However, the mechanisms of these effects are largely unknown. The purpose of this study is to elucidate NIR-induced molecular mechanisms on macrophages because macrophages play initial roles in directing immune responses by their M1 or M2 polarizations. Proteomic analysis revealed that NIR radiation enhanced the expression of mitochondrial respiratory gene citrate synthase. This increased citrate synthase expression was triggered by NIR-induced H3K4 hypermethylation on the citrate synthase gene promoter but not by heat, which led to macrophage M2 polarization and finally resulted in TGFß1 release from CD4+ cells. These cellular effects were validated in human primary macrophages and abdominal NIR-irradiated mouse experiments. In a phorbol 12-myristate 13-acetate‒induced inflammatory model on mouse ear, we confirmed that NIR irradiation induced significant anti-inflammatory effects through decreased M1 counts, reduced TNF-α, and increased CCL22 and/or TGFß1 levels.


Subject(s)
Dermatitis/therapy , Infrared Rays/therapeutic use , Macrophages/immunology , Phototherapy/methods , Animals , Citrate (si)-Synthase/metabolism , Dermatitis/immunology , Dermis/cytology , Dermis/immunology , Dermis/metabolism , Dermis/radiation effects , Disease Models, Animal , Female , Humans , Macrophage Activation/radiation effects , Macrophages/cytology , Macrophages/metabolism , Macrophages/radiation effects , Mice , Mitochondria/enzymology , Mitochondria/radiation effects , Primary Cell Culture , THP-1 Cells
5.
Nat Commun ; 11(1): 4064, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792542

ABSTRACT

Regulation of the programming of tumour-associated macrophages (TAMs) controls tumour growth and anti-tumour immunity. We examined the role of FGF2 in that regulation. Tumours in mice genetically deficient in low-molecular weight FGF2 (FGF2LMW) regress dependent on T cells. Yet, TAMS not T cells express FGF receptors. Bone marrow derived-macrophages from Fgf2LMW-/- mice co-injected with cancer cells reduce tumour growth and express more inflammatory cytokines. FGF2 is induced in the tumour microenvironment following fractionated radiation in murine tumours consistent with clinical reports. Combination treatment of in vivo tumours with fractionated radiation and a blocking antibody to FGF2 prolongs tumour growth delay, increases long-term survival and leads to a higher iNOS+/CD206+ TAM ratio compared to irradiation alone. These studies show for the first time that FGF2 affects macrophage programming and is a critical regulator of immunity in the tumour microenvironment.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Radiotherapy/methods , Animals , Cell Line, Tumor , Fibroblast Growth Factor 2/genetics , HT29 Cells , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophage Activation/drug effects , Macrophage Activation/radiation effects , Mannose Receptor , Mannose-Binding Lectins/genetics , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/radiation effects , Xenograft Model Antitumor Assays
6.
Lasers Med Sci ; 35(7): 1509-1518, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32065300

ABSTRACT

In spinal cord injury (SCI), inflammation is a major mediator of damage and loss of function and is regulated primarily by the bone marrow-derived macrophages (BMDMs). Photobiomodulation (PBM) or low-level light stimulation is known to have anti-inflammatory effects and has previously been used in the treatment of SCI, although its precise cellular mechanisms remain unclear. In the present study, the effect of PBM at 810 nm on classically activated BMDMs was evaluated to investigate the mechanisms underlying its anti-inflammatory effects. BMDMs were cultured and irradiated (810 nm, 2 mW/cm2) following stimulation with lipopolysaccharide and interferon-γ. CCK-8 assay, 2',7'-dichlorofluorescein diacetate assay, and ELISA and western blot analysis were performed to measure cell viability, reactive oxygen species production, and inflammatory marker production, respectively. PBM irradiation of classically activated macrophages significantly increased the cell viability and inhibited reactive oxygen species generation. PBM suppressed the expression of a marker of classically activated macrophages, inducible nitric oxide synthase; decreased the mRNA expression and secretion of pro-inflammatory cytokines, tumor necrosis factor alpha, and interleukin-1 beta; and increased the secretion of monocyte chemotactic protein 1. Exposure to PBM likewise significantly reduced the expression and phosphorylation of NF-κB p65 in classically activated BMDMs. Taken together, these results suggest that PBM can successfully modulate inflammation and polarization in classically activated BMDMs. The present study provides a theoretical basis to support wider clinical application of PBM in the treatment of SCI.


Subject(s)
Cell Polarity , Inflammation/radiotherapy , Macrophages/pathology , Animals , Cell Polarity/radiation effects , Cell Survival/radiation effects , Chemokines/genetics , Chemokines/metabolism , Gene Expression Regulation/radiation effects , Macrophage Activation/radiation effects , Macrophages/radiation effects , Mice, Inbred BALB C , Phosphorylation/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor RelA/metabolism
7.
Photochem Photobiol ; 96(4): 906-916, 2020 07.
Article in English | MEDLINE | ID: mdl-31907936

ABSTRACT

Moderate levels of a proinflammatory macrophages phenotype are indispensable and play an important role in the skeletal muscle repair process since this response depends on their secreted products concentration to influence and modulate muscle inflammation as well as the differentiation of myoblasts. This study investigated the effects of photobiomodulation (PBM) on undifferentiated and differentiation-induced C2C12 myoblasts cultivated in different concentrations of M1 phenotype macrophage-conditioned media of J774 cells (MCM1) also submitted to PBM using the same irradiation parameters. Irradiation was performed once with low-level laser (780 nm, 70 mW, 1 J) and was evaluated cell viability, proliferation and differentiation, nitric oxide (NO) synthesis and IL-6 and TNF-α protein levels 24 and 48 h after C2C12 irradiation. PBM treatment in undifferentiated myoblasts exhibited lower IL-6 levels in the presence of nonirradiated MCM1 at both concentrations. Myoblasts in proliferation condition cultivated with irradiated MCM1 showed lower IL-6 and TNF-α levels after 48 h in the presence of both concentrations evaluated. PBM induced a decrease in the synthesis of NO on undifferentiated and differentiation-induced myoblasts. PBM was able to reduce the level of proinflammatory protein and markers, which are important to allow the differentiation of myoblasts during the muscle repair process.


Subject(s)
Light , Macrophages/radiation effects , Myoblasts/radiation effects , Animals , Cell Line , Cell Proliferation/radiation effects , Culture Media, Conditioned , Macrophage Activation/radiation effects , Macrophages/cytology , Macrophages/metabolism , Mice , Nitric Oxide/biosynthesis
8.
J Immunol Res ; 2019: 3161750, 2019.
Article in English | MEDLINE | ID: mdl-31485459

ABSTRACT

Rheumatoid arthritis (RA) is a multifactorial autoimmune disease whose main hallmark is inflammation and destruction of the joints. Two cell types within the synovium that play an important role in RA are fibroblast-like synoviocytes (FLS) and macrophages. The latter innate immune cells show a high plasticity in their phenotype and are central in inflammatory processes. Low-dose radiotherapy (LD-RT) with particularly a single dose of 0.5 Gy has been demonstrated to have a positive impact on pain, inflammation, and bone in inflamed joints. We now examined for the first time how LD-RT influences FLS and bone marrow-derived macrophages in co-culture systems of an experimental model of RA to reveal further mechanisms of immune modulatory effects of low and intermediate dose of ionizing radiation. For this, the bone marrow of hTNF-α tg mice was differentiated either with cytokines to obtain key macrophage phenotypes (M0, M1, and M2) or with supernatants (SN) of untreated or irradiated FLS. Flow cytometry analyses were used to analyse the impact of radiation (0.1, 0.5, 1.0, and 2.0 Gy) on the phenotype of macrophages in the presence or absence of SN of FLS. LD-RT had no impact on cytokine-mediated macrophage polarization in M0, M1, or M2 macrophages. However, SN of irradiated FLS particularly reduced CD206 expression on macrophages. Macrophage phenotype was stable when being in contact with SN of nonirradiated FLS, but significantly increased surface expression of CD206 and slightly decreased CD80 and CD86 expression were observed when macrophage themselves were irradiated with 0.5 Gy under these microenvironmental conditions, again highlighting discontinuous dose dependencies in the low and intermediate dose range. One can conclude that FLS-dependent microenvironmental conditions have a slight influence on the modulation of macrophage phenotype under radiation exposure conditions. Future studies are needed to reveal the impact of radiation exposure on the functions of treated macrophages under such microenvironmental conditions.


Subject(s)
Arthritis, Rheumatoid/radiotherapy , Macrophages/radiation effects , Synoviocytes/radiation effects , Animals , Arthritis, Rheumatoid/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/immunology , Lectins, C-Type/metabolism , Macrophage Activation/radiation effects , Macrophages/immunology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Mice, Transgenic , Phenotype , Radiation Dosage , Receptors, Cell Surface/metabolism , Synoviocytes/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Technol Cancer Res Treat ; 18: 1533033819868225, 2019 01 01.
Article in English | MEDLINE | ID: mdl-31401938

ABSTRACT

OBJECTIVE: Tumor-treating fields are currently used to successfully treat various cancers; however, the specific pathways associated with its efficacy remain unknown in the immune responses. Here, we evaluated tumor-treating fields-mediated initiation of the macrophage-specific immune response. MATERIALS AND METHODS: We subjected RAW 264.7 mouse macrophages to clinically relevant levels of tumor-treating fields (0.9 V/cm, 150 kHz) and evaluated alterations in cytokine expression and release, as well as cell viability. Additionally, we investigated the status of immunomodulatory pathways to determine their roles in tumor-treating fields-mediated immune activation. RESULTS AND DISCUSSION: Our results indicated that tumor-treating fields treatment at 0.9 V/cm decreased cell viability and increased cytokine messenger RNA/protein levels, as well as levels of nitric oxide and reactive oxygen species, relative to controls. The levels of tumor necrosis factor α, interleukin 1ß, and interleukin 6 were markedly increased in tumor-treating fields-treated RAW 264.7 cells cocultured with 4T1 murine mammary carcinoma cells compared with those in 4T1 or RAW 264.7 cells with or without tumor-treating fields treatment. Moreover, the viability of 4T1 cells treated with the conditioned medium of tumor-treating fields-stimulated RAW 264.7 cells decreased, indicating that macrophage activation by tumor-treating fields effectively killed the tumor cells. Moreover, tumor-treating fields treatment activated the nuclear factor κB and mitogen-activated protein kinase pathways involved in immunomodulatory signaling. CONCLUSION: These results provide critical insights into the mechanisms through which tumor-treating fields affect macrophage-specific immune responses and the efficacy of this method for cancer treatment.


Subject(s)
Histocompatibility Antigens Class II/immunology , Macrophage Activation/immunology , Magnetic Field Therapy , Neoplasms/radiotherapy , Animals , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/radiation effects , Humans , MAP Kinase Signaling System/radiation effects , Macrophage Activation/genetics , Macrophage Activation/radiation effects , Macrophages/immunology , Macrophages/radiation effects , Mice , NF-kappa B/genetics , Neoplasms/immunology , Neoplasms/pathology , RAW 264.7 Cells , Signal Transduction/immunology , Signal Transduction/radiation effects
10.
Photobiomodul Photomed Laser Surg ; 37(5): 298-304, 2019 May.
Article in English | MEDLINE | ID: mdl-31084559

ABSTRACT

Objective: In this study, we evaluated the effectiveness of photodynamic therapy (PDT) for the treatment of experimental cutaneous leishmaniasis (CL) and the profile of macrophages activation markers. Background: Leishmaniasis is an infectious disease caused by parasites of the genus Leishmania. CL is caused by Leishmania major in the old world and by Leishmania braziliensis in the Americas. Considering the targeted organs, PDT may constitute a valuable therapeutic intervention. Macrophages are the host cells of Leishmania in mammals and may be classified into type M1 or M2 depending on the pattern of activation. Methods: BALB/c mice were infected in the foot pad with 1 × 106 amastigotes of L. braziliensis and treated with 5-aminolevulinic acid (5-ALA), visible light, or 5-ALA-PDT. The ex vivo mRNA expression levels of interleukin-10, tumor necrosis factor-α (TNF-α), arginase-1, heme oxygenase ( Hmox), and induced nitric oxide synthase (iNOS) were quantities as markers of macrophage activation with distinct ability to kill intracellular parasite. Results: The parasite load decreased significantly in the group treated with PDT compared with the other groups. The iNOS relative mRNA was higher in the group treated with PDT and light only compared with the group without treatment, whereas iNOS/arginase ratio was significantly higher only in the PDT group. The expression of TNF-α was significantly higher in 5-ALA and light compared with PDT and control group. No significant difference was observed in the expression of the other markers evaluated. Conclusions: Both, light and 5-ALA-PDT were able to upregulate iNOS expression only; 5-ALA-PDT was able to reduce parasite burden. The increase in the iNOS levels suggests it might participate in the antimicrobial mechanisms triggered by 5-ALA-PDT; although parasite death mechanism was not completely clarified, the results presented in this study suggest that macrophage activation may contribute to parasite control.


Subject(s)
Aminolevulinic Acid/therapeutic use , Leishmaniasis, Cutaneous/therapy , Macrophage Activation/radiation effects , Photochemotherapy , Photosensitizing Agents/therapeutic use , Animals , Disease Models, Animal , Interleukin-10/metabolism , Leishmaniasis, Cutaneous/metabolism , Leishmaniasis, Cutaneous/pathology , Male , Mice , Mice, Inbred BALB C , Parasite Load , Tumor Necrosis Factor-alpha/metabolism
11.
Gut ; 68(6): 1003-1013, 2019 06.
Article in English | MEDLINE | ID: mdl-29934438

ABSTRACT

OBJECTIVE: Lactobacillus rhamnosus GG (LGG), a probiotic, given by gavage is radioprotective of the mouse intestine. LGG-induced radioprotection is toll-like receptor 2 (TLR2) and cyclooxygenase-2 (COX-2)-dependent and is associated with the migration of COX-2+mesenchymal stem cells (MSCs) from the lamina propria of the villus to the lamina propria near the crypt epithelial stem cells. Our goals were to define the mechanism of LGG radioprotection including identification of the TLR2 agonist, and the mechanism of the MSC migration and to determine the safety and efficacy of this approach in models relevant to clinical radiation therapy. DESIGN: Intestinal radioprotection was modelled in vitro with cell lines and enteroids as well as in vivo by assaying clinical outcomes and crypt survival. Fractionated abdominal and single dose radiation were used along with syngeneic CT26 colon tumour grafts to assess tumour radioprotection. RESULTS: LGG with a mutation in the processing of lipoteichoic acid (LTA), a TLR2 agonist, was not radioprotective, while LTA agonist and native LGG were. An agonist of CXCR4 blocked LGG-induced MSC migration and LGG-induced radioprotection. LGG given by gavage induced expression of CXCL12, a CXCR4 agonist, in pericryptal macrophages and depletion of macrophages by clodronate liposomes blocked LGG-induced MSC migration and radioprotection. LTA effectively protected the normal intestinal crypt, but not tumours in fractionated radiation regimens. CONCLUSIONS: LGG acts as a 'time-release capsule' releasing radioprotective LTA. LTA then primes the epithelial stem cell niche to protect epithelial stem cells by triggering a multicellular, adaptive immune signalling cascade involving macrophages and PGE2 secreting MSCs. TRIAL REGISTRATION NUMBER: NCT01790035; Pre-results.


Subject(s)
Intestinal Mucosa/metabolism , Lacticaseibacillus rhamnosus , Lipopolysaccharides/metabolism , Probiotics/pharmacology , Radiation Injuries/prevention & control , Teichoic Acids/metabolism , Animals , Cell Movement/radiation effects , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Macrophage Activation/radiation effects , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Radiation-Protective Agents , Reference Values , Sensitivity and Specificity
12.
Cell Death Differ ; 24(9): 1632-1644, 2017 09.
Article in English | MEDLINE | ID: mdl-28574504

ABSTRACT

Although tumor-associated macrophages have been extensively studied in the control of response to radiotherapy, the molecular mechanisms involved in the ionizing radiation-mediated activation of macrophages remain elusive. Here we show that ionizing radiation induces the expression of interferon regulatory factor 5 (IRF5) promoting thus macrophage activation toward a pro-inflammatory phenotype. We reveal that the activation of the ataxia telangiectasia mutated (ATM) kinase is required for ionizing radiation-elicited macrophage activation, but also for macrophage reprogramming after treatments with γ-interferon, lipopolysaccharide or chemotherapeutic agent (such as cisplatin), underscoring the fact that the kinase ATM plays a central role during macrophage phenotypic switching toward a pro-inflammatory phenotype through the regulation of mRNA level and post-translational modifications of IRF5. We further demonstrate that NADPH oxidase 2 (NOX2)-dependent ROS production is upstream to ATM activation and is essential during this process. We also report that the inhibition of any component of this signaling pathway (NOX2, ROS and ATM) impairs pro-inflammatory activation of macrophages and predicts a poor tumor response to preoperative radiotherapy in locally advanced rectal cancer. Altogether, our results identify a novel signaling pathway involved in macrophage activation that may enhance the effectiveness of radiotherapy through the reprogramming of tumor-infiltrating macrophages.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Macrophage Activation/radiation effects , Macrophages/metabolism , Animals , Cell Line , Flow Cytometry , Humans , Interferon-gamma/metabolism , Mice , Microscopy, Fluorescence , Phosphorylation/radiation effects , Protein Processing, Post-Translational , RAW 264.7 Cells , Signal Transduction
13.
Radiat Res ; 187(5): 549-561, 2017 05.
Article in English | MEDLINE | ID: mdl-28319461

ABSTRACT

In patients undergoing radiotherapy for cancer, radiation dose to healthy tissue can occur, causing microvascular damage. Monocytes that have been shown to promote tissue revascularization comprise the subsets: CD11b+Ly6G-7/4hi/monocyteshi and CD11b+Ly6G-7/4lo/monocyteslo. We hypothesized that monocytes were implicated in postirradiation blood vessel formation. C57Bl6 mice underwent localized colorectal irradiation and were sacrificed at different times after exposure. Bone marrow, spleen, blood and colon were collected. Fourteen days postirradiation, colons expressed proangiogenic actors and adhesion molecules. Monocyteshi, which were the main subset of infiltrating monocytes, mobilized to the blood from spleen and bone marrow, peaking at day 14 postirradiation, and were associated with lymphocyte Th1 polarization. At day 28 postirradiation, angiographic score and capillary density increased by ∼1.8-fold, and then returned to nonirradiated levels at day 60. Clodronate-mediated depletion of circulating monocytes prior to irradiation resulted in a ∼1.4-fold decrease in angiographic score and capillary density compared to the nontreated control. Histological analysis of the colon in clodronate-treated mice revealed a massive decrease of macrophage and lymphocyte infiltration as well as reduced collagen deposition in crypt area at day 21. However, late depletion of monocytes from day 25 postirradiation had no effect on fibrotic process. These findings demonstrate a central role for monocyte/macrophage activation in the orchestration of a neovascularization mechanism after localized colorectal irradiation.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/radiotherapy , Macrophages/immunology , Monocytes/immunology , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/immunology , Animals , Cell Movement/immunology , Cell Movement/radiation effects , Colorectal Neoplasms/complications , Female , Macrophage Activation/immunology , Macrophage Activation/radiation effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Monocytes/radiation effects , Neovascularization, Pathologic/radiotherapy , Radiotherapy Dosage , Radiotherapy, Conformal/adverse effects , Treatment Outcome
14.
Int J Biol Macromol ; 97: 181-189, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28082222

ABSTRACT

Scavenger receptors (SRs) expressed on the activated macrophages in inflammation sites have been considered as the most interesting and important target biomarker for targeted drug delivery, imaging and therapy. In the present study, we fabricated the scavenger receptor-A (SR-A) targeted-photoactivatable nanoagents (termed as Ce6/DS-DOCA) by entrapping chlorin e6 (Ce6) into the amphiphilic dextran sulfate-deoxycholic acid (DS-DOCA) conjugates via physically hydrophobic interactions. Insoluble Ce6 was easily encapsulated into DS-DOCA nanoparticles by a dialysis method and the loading efficiency was approximately 51.7%. The Ce6/DS-DOCA formed nano-sized self-assembled aggregates (28.8±5.6nm in diameter), confirmed by transmission electron microscope, UV/Vis and fluorescence spectrophotometer. The Ce6/DS-DOCA nanoagents could generate highly reactive singlet oxygen under laser irradiation. Also, in vitro studies showed that they were more specifically taken up by lipopolysaccharide (LPS)-induced activated macrophages (RAW 264.7) via a SR-A-mediated endocytosis, relative to by non-activated macrophages, and notably induced cell death of activated macrophages under laser irradiation. Therefore, SR-A targetable and photoactivatable Ce6/DS-DOCA nanoagents with more selective targeting to the activated macrophages will have great potential for treatment of inflammatory diseases.


Subject(s)
Macrophage Activation/drug effects , Macrophages/drug effects , Molecular Targeted Therapy , Nanoparticles/chemistry , Photochemotherapy , Porphyrins/pharmacology , Scavenger Receptors, Class A/metabolism , Animals , Biological Transport , Chlorophyllides , Deoxycholic Acid/chemistry , Dextran Sulfate/chemistry , Drug Carriers/chemistry , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Hydrophobic and Hydrophilic Interactions , Intracellular Space/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/radiation effects , Macrophages/immunology , Macrophages/radiation effects , Mice , Porphyrins/chemistry , Porphyrins/metabolism , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scavenger Receptors, Class A/genetics , Solubility
15.
Lasers Med Sci ; 31(8): 1613-1619, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27475996

ABSTRACT

Effective decontamination of biofilm and bacterial toxins from the surface of dental implants is a yet unresolved issue. This in vitro study aims at providing the experimental basis for possible use of diode laser (λ 808 nm) in the treatment of peri-implantitis. Staphylococcus aureus biofilm was grown for 48 h on titanium discs with porous surface corresponding to the bone-implant interface and then irradiated with a diode laser (λ 808 nm) in noncontact mode with airflow cooling for 1 min using a Ø 600-µm fiber. Setting parameters were 2 W (400 J/cm2) for continuous wave mode; 22 µJ, 20 kHz, 7 µs (88 J/cm2) for pulsed wave mode. Bactericidal effect was evaluated using fluorescence microscopy and counting the residual colony-forming units. Biofilm and titanium surface morphology were analyzed by scanning electron microscopy (SEM). In parallel experiments, the titanium discs were coated with Escherichia coli lipopolysaccharide (LPS), laser-irradiated and seeded with RAW 264.7 macrophages to quantify LPS-driven inflammatory cell activation by measuring the enhanced generation of nitric oxide (NO). Diode laser irradiation in both continuous and pulsed modes induced a statistically significant reduction of viable bacteria and nitrite levels. These results indicate that in addition to its bactericidal effect laser irradiation can also inhibit LPS-induced macrophage activation and thus blunt the inflammatory response. The λ 808-nm diode laser emerges as a valuable tool for decontamination/detoxification of the titanium implant surface and may be used in the treatment of peri-implantitis.


Subject(s)
Bacterial Adhesion/radiation effects , Biofilms/drug effects , Dental Implants/microbiology , Escherichia coli/chemistry , Lasers, Semiconductor , Lipopolysaccharides/pharmacology , Staphylococcus aureus/radiation effects , Titanium/pharmacology , Animals , Decontamination , Fluorescence , Macrophage Activation/drug effects , Macrophage Activation/radiation effects , Mice , Microbial Viability/radiation effects , RAW 264.7 Cells , Staphylococcus aureus/drug effects , Staphylococcus aureus/ultrastructure , Surface Properties
16.
Int J Radiat Biol ; 92(12): 754-765, 2016 12.
Article in English | MEDLINE | ID: mdl-27539247

ABSTRACT

PURPOSE: Thoracic irradiation injures lung parenchyma, triggering inflammation and immune cell activation, leading to pneumonitis and fibrosis. Macrophage polarization contributes to these processes. Since IL-4 promotes pro-fibrotic macrophage activation, its role in radiation-induced lung injury was investigated. MATERIALS AND METHODS: Lung macrophage subpopulations were characterized from 3-26 weeks following exposure of WT and IL-4-/- mice to 0 or 12.5 Gray single dose thoracic irradiation. RESULTS: Loss of IL-4 did not prevent fibrosis, but blunted macrophage accumulation within the parenchyma. At 3 weeks following exposure, cell numbers and expression of F4/80 and CD206, an alternative activation marker, decreased in alveolar macrophages but increased in infiltrating macrophages in WT mice. Loss of IL-4 impaired recovery of these markers in alveolar macrophages and blunted expansion of these populations in infiltrating macrophages. CD206+ cells were evident in fibrotic regions of WT mice only, however Arg-1+ cells increased in fibrotic regions in IL-4-/- mice only. Radiation-induced proinflammatory Ly6C expression was more apparent in alveolar and interstitial macrophages from IL-4-/- mice. CONCLUSIONS: IL-4 loss did not prevent alternative macrophage activation and fibrosis in irradiated mice. Instead, a role is indicated for IL-4 in maintenance of macrophage populations in the lung following high single dose thoracic irradiation.


Subject(s)
Interleukin-4/immunology , Macrophages/immunology , Macrophages/radiation effects , Pulmonary Fibrosis/immunology , Radiation Exposure/adverse effects , Radiation Pneumonitis/immunology , Animals , Dose-Response Relationship, Radiation , Female , Lung , Macrophage Activation/immunology , Macrophage Activation/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Fibrosis/etiology , Radiation Dosage , Radiation Pneumonitis/etiology
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 32(8): 1045-50, 2016 Aug.
Article in Chinese | MEDLINE | ID: mdl-27412934

ABSTRACT

Objective To investigate the influence of 810 nm low-level laser of different energy on the polarization of macrophages. Methods The macrophages were isolated from the bone borrow of BALB/c mice and cultured in macrophage colony stimulating factor (M-CSF) conditioned cultural medium. The expression of F4/80 was examined by flow cytometry for identification. After lipopolysaccharide-γ interferon (LPS-IFN-γ) induced polarization status in the macrophages, the mRNA expressions of inducible nitric oxide synthase (iNOS), arginase 1 (Arg1) and CD86 were detected by reverse transcription PCR, and the protein expressions of iNOS and Arg1 were tested by Western blotting. Thereafter, the M1 macrophages were exposed to 810 nm low-level laser of (1, 2, 3, 4) J/cm(2), and then the cell viability was evaluated by MTT assay; the expressions of iNOS and Arg1 were observed by immunofluorescent cytochemical staining; the mRNA and protein levels of iNOS and Arg1 were studied by reverse transcription PCR and Western blotting. Results Flow cytometry showed that the percentage of F4/80 positive cells cultured with M-CSF conditioned medium was 99.9%. The mRNA and protein levels of iNOS and CD86 in macrophages were both significantly raised after induction by LPS-IFN-γ. Compared with the control cells, the viability of M1 cells significantly decreased when the energy of the low-level laser exposure was 4 J/cm(2), while the viability remained unchanged when the energy was 1, 2 or 3 J/cm(2). Immunocytochemistry revealed that the percentage of Arg1 positive cells that represent M2 macrophages was not significantly different from the control group when the irradiation dose was 1 or 2 J/cm(2), however, the Arg1 positive cells significantly increased and the iNOS positive cells that represent M1 macrophages significantly decreased when the irradiation dose was 3 or 4 J/cm(2). When the irradiation dose was 1 or 2 J/cm(2), the mRNA and protein levels of iNOS and Arg1 remained unchanged compared with the control group. When the irradiation dose was 3 or 4 J/cm(2), the mRNA and protein levels of iNOS significantly decreased and Arg1 significantly increased. Conclusion The 810 nm low-level laser with the energy of 1 or 2 J/cm(2) have no significant influence on the polarization and viability of macrophages. M1 macrophages can be polarized into M2 macrophages while there is no significant difference in the cell viability when the energy was 3 J/cm(2). When the energy was 4 J/cm(2), M1 cells can be polarized into M2 cells and the cell viability significantly decreased.


Subject(s)
Gene Expression/radiation effects , Lasers , Macrophage Activation/radiation effects , Macrophages/radiation effects , Animals , Animals, Newborn , Arginase/genetics , Arginase/metabolism , Blotting, Western , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Gene Expression/drug effects , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Reverse Transcriptase Polymerase Chain Reaction
18.
Antioxid Redox Signal ; 24(5): 249-62, 2016 Feb 10.
Article in English | MEDLINE | ID: mdl-26413929

ABSTRACT

AIMS: Our previous studies have demonstrated that as a mitochondria-targeting cancer phototherapy, high-fluence, low-power laser irradiation (HF-LPLI) results in oxidative damage that induces tumor cell apoptosis. In this study, we focused on the immunological effects of HF-LPLI phototherapy and explored its antitumor immune regulatory mechanism. RESULTS: We found not only that HF-LPLI treatment induced tumor cell apoptosis but also that HF-LPLI-treated apoptotic tumor cells activated macrophages. Due to mitochondrial superoxide anion burst after HF-LPLI treatment, tumor cells displayed a high level of phosphatidylserine oxidation, which mediated the recognition and uptake by macrophages with the subsequent secretion of cytokines and generation of cytotoxic T lymphocytes. In addition, in vivo results showed that HF-LPLI treatment caused leukocyte infiltration into the tumor and efficaciously inhibited tumor growth in an EMT6 tumor model. These phenomena were absent in the respiration-deficient EMT6 tumor model, implying that the HF-LPLI-elicited immunological effects were dependent on the mitochondrial superoxide anion burst. INNOVATION: In this study, for the first time, we show that HF-LPLI mediates tumor-killing effects via targeting photoinactivation of respiratory chain oxidase to trigger a superoxide anion burst, leading to a high level of oxidatively modified moieties, which contributes to the phenotypic changes in macrophages and mediates the antitumor immune response. CONCLUSION: Our results suggest that HF-LPLI may be an effective cancer treatment modality that both eradicates the treated primary tumors and induces an antitumor immune response via photoinactivation of respiratory chain oxidase to trigger superoxide anion burst.


Subject(s)
Neoplasms/immunology , Neoplasms/metabolism , Phototherapy , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Cytokines/biosynthesis , Disease Models, Animal , Electron Transport , Female , Humans , Low-Level Light Therapy/methods , Macrophage Activation/immunology , Macrophage Activation/radiation effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/radiation effects , Mice , Mitochondria/metabolism , Mitochondria/radiation effects , NADPH Oxidases/metabolism , Neoplasms/pathology , Neoplasms/therapy , Oxidation-Reduction/radiation effects , Phototherapy/methods , Reactive Oxygen Species/metabolism , Superoxides/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/radiation effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
19.
Lasers Med Sci ; 31(2): 289-96, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26714983

ABSTRACT

The aim of the present study was to investigate the therapeutic mechanism of low-level laser therapy (LLLT) in the mouse tail lymphedema model. Six-week-old female mice were classified into the laser treatment group, sham treatment group, and surgical control group (10 mice per group). LLLT was administered daily for 10 min from the surgical day to 11 days (12 times). Macrophage activation and lymphatic vessel regeneration were evaluated through immunohistochemical staining with anti-F4/80 and anti-LYVE-1 antibodies, respectively, at 12 days post-procedure. Quantitative real-time polymerase chain reaction (qPCR) was performed to measure messenger RNA (mRNA) expression of vascular endothelial growth factor A, B, C, R1, R2, and R3 (VEGF-A, VEGF-B, VEGF-C, VEGFR1, VEGFR2, and VEGFR3) at 12 days post-procedure. Student's t and one-way ANOVA tests were performed for statistical analyses. Significance was defined as p < 0.05. The thickness of the tail rapidly increased until 6 days in the laser and sham groups. The mice in the laser group showed a significantly decreased thickness compared with the sham group at 10 and 12 days. Immunohistochemistry assay revealed that LLLT reduced inflammation and induced new lymphatic vessel growth. qPCR showed that expressions of VEGFR3 were (p = 0.002) increased in the laser group. These results suggest that LLLT has anti-inflammatory and lymphangiogenetic effects for the management of lymphedema.


Subject(s)
Low-Level Light Therapy , Lymphangiogenesis/radiation effects , Lymphedema/radiotherapy , Animals , Disease Models, Animal , Female , Gene Expression Regulation/radiation effects , Inflammation/radiotherapy , Lymphatic Vessels/physiopathology , Lymphatic Vessels/radiation effects , Lymphedema/genetics , Lymphedema/immunology , Lymphedema/physiopathology , Macrophage Activation/radiation effects , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Regeneration/radiation effects , Vascular Endothelial Growth Factor A/genetics
20.
J Thorac Oncol ; 10(12): 1703-12, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26709479

ABSTRACT

INTRODUCTION: Radiation therapy for lung cancer is limited by toxicity to normal lung tissue that results from an inflammatory process, leading to pneumonitis and fibrosis. Soy isoflavones mitigate inflammatory infiltrates and radiation-induced lung injury, but the cellular immune mediators involved in the radioprotective effect are unknown. METHODS: Mice received a single dose of 10 Gy radiation delivered to the lungs and daily oral treatment of soy isoflavones. At different time points, mice were either processed to harvest bronchoalveolar lavage fluid for differential cell counting and lungs for flow cytometry or immunohistochemistry studies. RESULTS: Combined soy and radiation led to a reduction in infiltration and activation of alveolar macrophages and neutrophils in both the bronchoalveolar and lung parenchyma compartments. Soy treatment protected F4/80CD11c interstitial macrophages, which are known to play an immunoregulatory role and are decreased by radiation. Furthermore, soy isoflavones reduced the levels of nitric oxide synthase 2 expression while increasing arginase-1 expression after radiation, suggesting a switch from proinflammatory M1 macrophage to an anti-inflammatory M2 macrophage phenotype. Soy also prevented the influx of activated neutrophils in lung caused by radiation. CONCLUSIONS: Soy isoflavones inhibit the infiltration and activation of macrophages and neutrophils induced by radiation in lungs. Soy isoflavones-mediated modulation of macrophage and neutrophil responses to radiation may contribute to a mechanism of resolution of radiation-induced chronic inflammation leading to radioprotection of lung tissue.


Subject(s)
Isoflavones/pharmacology , Lung Neoplasms/radiotherapy , Lung/drug effects , Lung/radiation effects , Macrophages/drug effects , Radiation Injuries, Experimental/prevention & control , Radiation-Protective Agents/pharmacology , Animals , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Macrophage Activation/drug effects , Macrophage Activation/radiation effects , Macrophages/metabolism , Macrophages/pathology , Macrophages/radiation effects , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Neutrophils/radiation effects , Radiation Injuries, Experimental/metabolism , Glycine max/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL