Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Neoplasia ; 46: 100940, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37913654

ABSTRACT

Radiation therapy is an established and effective anti-cancer treatment modality. Extensive pre-clinical experimentation has demonstrated that the pro-inflammatory properties of irradiation may be synergistic with checkpoint immunotherapy. Radiation induces double-stranded DNA breaks (dsDNA). Sensing of the dsDNA activates the cGAS/STING pathway, producing Type 1 interferons essential to recruiting antigen-presenting cells (APCs). Radiation promotes cytotoxic CD8 T-cell recruitment by releasing tumour-associated antigens captured and cross-presented by surveying antigen-presenting cells. Radiation-induced vascular normalisation may further promote T-cell trafficking and drug delivery. Radiation is also immunosuppressive. Recruitment of regulatory T cells (Tregs) and innate cells such as myeloid-derived suppressive cells (m-MDSCs) all counteract the immunostimulatory properties of radiation. Many innate immune cell types operate at the interface of the adaptive immune response. Innate immune cells, such as m-MDSCs, can exert their immunosuppressive effects by expressing immune checkpoints such as PD-L1, further highlighting the potential of combined radiation and checkpoint immunotherapy. Several early-phase clinical studies investigating the combination of radiation and immunotherapy have been disappointing. A greater appreciation of radiotherapy's impact on the innate immune system is essential to optimise radioimmunotherapy combinations. This review will summarise the impact of radiotherapy on crucial cells of the innate immune system and vital immunosuppressive cytokines.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Immunity, Innate , Neoplasms/radiotherapy , Adaptive Immunity/radiation effects , Antineoplastic Agents/pharmacology , Immunotherapy , Tumor Microenvironment
2.
J Cancer Res Ther ; 17(2): 537-542, 2021.
Article in English | MEDLINE | ID: mdl-34121704

ABSTRACT

INTRODUCTION: In the present study, the radioadaptive role of the immune system induced by low dose (LD) was investigated for its in vivo protective activity. MATERIALS AND METHODS: Quantitative analysis of cytokine gene expression was assessed for their in vivo activity in BALB/c mice. To evaluate the adaptive response induced by LD on the mice spleen lymphocyte, the cytokine interleukin (IL)-4, interferon (IFN)-γ, and transforming growth factor (TGF)-ß expression was measured by a real-time quantitative polymerase chain reaction. To verify the radioadaptive effect of LD, animals were preirradiated at 10 cGy from a 60 Co source and then challenge dose at 200 cGy was delivered. Independent sample student's t-test was employed to compare cytokine gene expression in radioadaptive (10 + 200 cGy), LD (10 cGy), high-dose (HD, 200 cGy), and control groups of animals. RESULTS: Following the HD, the cytokine gene expression of IFN-γ, IL-4, and TGF-ß was significantly decreased compared to the control group (P = 0.0001). However, TGF-ß expression was also decreased significantly in the LD and adaptive groups compared to the control group (P = 0.0001). IFN-γ/IL-4 ratio in the adaptive group was significantly decreased compared to the HD group (P = 0.0001). CONCLUSION: These results indicate that the immune system plays an important role for radioadaptive response induction by LD radiation to adjust the harmful effects of HD irradiation.


Subject(s)
Adaptation, Physiological/immunology , Adaptive Immunity/radiation effects , Gene Expression Regulation/radiation effects , Adaptation, Physiological/radiation effects , Animals , Cells, Cultured , Dose-Response Relationship, Radiation , Gene Expression Regulation/immunology , Interferon-gamma/genetics , Interleukin-4/genetics , Male , Mice , Models, Animal , Primary Cell Culture , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Transforming Growth Factor beta/genetics , Whole-Body Irradiation
3.
Int J Radiat Oncol Biol Phys ; 111(2): 502-514, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34023423

ABSTRACT

PURPOSE: We examined how radiation dose per fraction (DPF) and total dose, as represented by biological effective dose (BED), can independently and differentially affect the immunomodulatory capacity of radiation therapy (RT). METHODS AND MATERIALS: AT3-OVA mammary and MC38 colorectal tumors in C57BL/6 mice were irradiated with rationally selected dose-fractionation schedules, alone or with immune-modulating or -depleting agents. Tumor growth was monitored as a readout of therapeutic response. Flow cytometry and RNA sequencing of mouse tumors and analysis of transcriptomic data sets from irradiated human cancers were used to examine the immunomodulatory effects of the different radiation schedules. RESULTS: In AT3-OVA tumors, radiation DPF rather than BED determined the ability of RT to evoke local antitumor CD8+ T cell responses and synergize with anti-PD-1 therapy. Natural killer cell-mediated control of irradiated tumors was more sensitive to radiation BED. Radiation-induced regulatory T cell (Treg) responses, which were detected in both mouse and human tumors, were a major factor underlying the differential activation of adaptive immunity by radiation DPF and the activity of natural killer cells during the early phase of response to RT. Targeted inhibition of Treg responses within irradiated tumors rescued and enhanced local tumor control by RT and permitted the generation of abscopal and immunologic memory responses, irrespective of radiation schedule. MC38 tumors did not support the induction of an amplified Treg response to RT and were highly vulnerable to its immunoadjuvant effects. CONCLUSIONS: Local radiation-induced Treg responses are influenced by radiation schedule and tumor type and are a critical determinant of the immunoadjuvant potential of RT and its ability to synergize with T cell-targeted immunotherapy.


Subject(s)
Dose Fractionation, Radiation , Neoplasms, Experimental/radiotherapy , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity/radiation effects , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Innate/radiation effects , Immunomodulation , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology
4.
Ital J Dermatol Venerol ; 156(3): 366-373, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33913665

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), has become pandemic on March 11th, 2020. COVID-19 has a range of symptoms that includes fever, fatigue, dry cough, aches, and labored breathing to acute respiratory distress and possibly death. Health systems and hospitals have been completely rearranged since March 2020 in order to limit the high rate of virus spreading. Hence, a great debate on deferrable visits and treatments including phototherapy for skin diseases is developing. In particular, as regards phototherapy very few data are currently available regarding the chance to continue it, even if it may be a useful resource for treating numerous dermatological patients. However, phototherapy has an immunosuppressive action possibly facilitating virus infection. In the context of COVID-19 infection risk it is important to pointed out whether sunlight, phototherapy and in particular ultraviolet radiation (UV-R) constitute or not a risk for patients. In this review we aimed to focus on the relationship between UV-R, sunlight, phototherapy, and viral infections particularly focusing on COVID-19.


Subject(s)
COVID-19/epidemiology , Pandemics , SARS-CoV-2/radiation effects , Sunlight , Ultraviolet Rays , Vitamin D/physiology , Adaptive Immunity/radiation effects , Animals , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/physiology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Humans , Immunity, Innate/radiation effects , Immunosuppression Therapy , Interleukin-6/blood , Pathogen-Associated Molecular Pattern Molecules , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Skin Diseases/radiotherapy , Sunlight/adverse effects , Toll-Like Receptors/physiology , Ultraviolet Rays/adverse effects , Ultraviolet Therapy/adverse effects , Viruses/radiation effects , Vitamin D/biosynthesis , Vitamin D/therapeutic use , Cathelicidins
5.
J Appl Toxicol ; 41(9): 1425-1437, 2021 09.
Article in English | MEDLINE | ID: mdl-33368402

ABSTRACT

Zinc oxide nanoparticles (ZnO-NPs) are widely used in almost every area of life. Therefore, exposure to them is unavoidable, which makes it necessary to assess their safety for humans. This paper aims to determine toxicity of ZnO-NPs of two diameters toward human immune cells responsible for: innate response (U-937 and HL-60) and acquired response (COLO-720L and HUT-78). Mitochondrial activity, membrane integrity, degree of cellular lipid oxidation, induction of inflammation, and activation of the apoptosis pathway were evaluated to determine differences in cellular response to the tested nanoparticles. ZnO-NPs with a diameter of 100 and 130 nm cause significant cell mortality at 25 and 12 mg/L, respectively. Mitochondrial damage leads to the activation of the caspase cascade and, consequently, to cell apoptosis. ZnO-NPs also cause peroxidation of membrane lipids. Due to the photocatalytic properties of ZnO-NPs, the effect of ultraviolet (UV) irradiation on the degree of their toxicity was also investigated. However, UV irradiation enhances the toxic effect of nanoparticles mainly by weakening the cell's defense capabilities. ZnO-NPs are cytotoxic to human immune system, and they may cause both long-term and short-term negative effects.


Subject(s)
Adaptive Immunity/drug effects , Immunity, Cellular/drug effects , Immunity, Innate/drug effects , Metal Nanoparticles/toxicity , Zinc Oxide/toxicity , Adaptive Immunity/radiation effects , Apoptosis/drug effects , Caspases/metabolism , Cell Line , Cell Membrane/drug effects , Cell Membrane/pathology , Humans , Immunity, Cellular/radiation effects , Immunity, Innate/radiation effects , Inflammation/chemically induced , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Particle Size , Reactive Oxygen Species , Signal Transduction/drug effects , Ultraviolet Rays
6.
Br J Cancer ; 123(3): 339-348, 2020 08.
Article in English | MEDLINE | ID: mdl-32581341

ABSTRACT

Radiotherapy is a pivotal component in the curative treatment of patients with localised cancer and isolated metastasis, as well as being used as a palliative strategy for patients with disseminated disease. The clinical efficacy of radiotherapy has traditionally been attributed to the local effects of ionising radiation, which induces cell death by directly and indirectly inducing DNA damage, but substantial work has uncovered an unexpected and dual relationship between tumour irradiation and the host immune system. In clinical practice, it is, therefore, tempting to tailor immunotherapies with radiotherapy in order to synergise innate and adaptive immunity against cancer cells, as well as to bypass immune tolerance and exhaustion, with the aim of facilitating tumour regression. However, our understanding of how radiation impacts on immune system activation is still in its early stages, and concerns and challenges regarding therapeutic applications still need to be overcome. With the increasing use of immunotherapy and its common combination with ionising radiation, this review briefly delineates current knowledge about the non-targeted effects of radiotherapy, and aims to provide insights, at the preclinical level, into the mechanisms that are involved with the potential to yield clinically relevant combinatorial approaches of radiotherapy and immunotherapy.


Subject(s)
Bystander Effect , Neoplasms/radiotherapy , Tumor Escape/radiation effects , Adaptive Immunity/radiation effects , Animals , Humans , Immunity, Innate/radiation effects , Neoplasms/immunology , Radioimmunotherapy
7.
Semin Radiat Oncol ; 30(2): 129-138, 2020 04.
Article in English | MEDLINE | ID: mdl-32381292

ABSTRACT

The anti-tumor activity of interferons (IFNs) was first appreciated about half a century ago, and IFN-α2 was the first cancer immunotherapy approved by the US Food and Drug Administration. Radiation therapy (RT), one of the pillars of cancer treatment, directly causes DNA damage, which can lead to senescence and cell death in tumor cells. In recent years, however, RT-induced immunomodulatory effects have been recognized to play an indispensable role in achieving the optimum therapeutic effect of RT. Increasing evidence indicates that RT enhances adaptive anti-tumor immunity by augmenting the innate immune sensing of tumors in a type I IFN-dependent matter. This review briefly introduces the role of type I interferon in cancer and the available evidence on the overall effects of RT on tumor immunity mediated via type I IFN. Recent advances in deciphering the molecular mechanisms underlying the induction of type I IFNs triggered by RT, their clinical implications, and therapeutic opportunities will be highlighted.


Subject(s)
Immunotherapy/methods , Interferon Type I/immunology , Neoplasms/immunology , Neoplasms/radiotherapy , Adaptive Immunity/radiation effects , Combined Modality Therapy , Humans , Immunity, Innate/radiation effects , Interferon Type I/pharmacology
8.
Semin Radiat Oncol ; 30(2): 145-157, 2020 04.
Article in English | MEDLINE | ID: mdl-32381294

ABSTRACT

A malignant tumor consists of malignant cells as well as a wide array of normal host tissues including stroma, vasculature, and immune infiltrate. The interaction between cancer and these host tissues is critical as these host tissues play a variety of roles in supporting or resisting disease progression. Radiotherapy (RT) has direct effects on malignant cells, but, also, critically important effects on these other components of the tumor microenvironment (TME). Given the growing role of immune checkpoint inhibitors and other immunotherapy strategies, understanding how RT affects the TME, particularly the immune compartment, is essential to advance RT in this new era of cancer therapy. The interactions between RT and the TME are complex, affecting the innate and adaptive arms of the immune system. RT can induce both proinflammatory effects and immune suppressive effects that can either promote or impede antitumor immunity. It is likely that the initial proinflammatory effects of RT eventually lead to rebound immune-suppression as chronic inflammation sets in. The exact kinetics and nature of how RT changes the TME likely depends on timing, dose, fractionation, site irradiated, and tumor type. With increased understanding of the effects of RT on the TME, in the future it is likely that we will be able to personalize RT by varying the dose, site, and timing of intervention to generate the desired response to partner with immunotherapy strategies.


Subject(s)
Immunotherapy/methods , Neoplasms/immunology , Neoplasms/radiotherapy , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects , Adaptive Immunity/radiation effects , Humans , Immunity, Innate/radiation effects
9.
Semin Radiat Oncol ; 30(2): 139-144, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32381293

ABSTRACT

The role of radiation therapy for cancer is evolving as its biologic effects on tumors becomes more clearly defined. Traditionally, radiobiology models dose-response curves based on direct cytocidal effects of radiation on tumor cells. However, a more dynamic picture is emerging of the impact of radiation on the tumor microenvironment and the patient's innate and adaptive immune system. Radiation produces damage associated molecular patterns that activate innate immune receptors leading to a cascade of downstream signals. These signals alter the tumor infiltrating immune population, modulate immune cell activation status, and enhance antigen presentation. The changes are conducive to an adaptive immune response with the generation of antitumor T cells. Early findings from clinical trials incorporating radiation and immune checkpoint inhibitors show promising synergy between the 2 modalities. As more data matures from ongoing trials, clinicians can refine new strategies for using radiation together with immunotherapy to achieve improved clinical outcomes.


Subject(s)
Adaptive Immunity/radiation effects , Immunity, Innate/radiation effects , Immunotherapy/methods , Neoplasms/immunology , Neoplasms/radiotherapy , Tumor Microenvironment/radiation effects , Combined Modality Therapy , Humans , Tumor Microenvironment/immunology
10.
Int J Radiat Oncol Biol Phys ; 108(1): 93-103, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32311417

ABSTRACT

PURPOSE: The role of MerTK, a member of the Tyro3-Axl-MerTK family of receptor tyrosine kinase, in the immune response to radiation therapy (RT) is unclear. We investigated immune-mediated tumor control after RT in murine models of colorectal and pancreatic adenocarcinoma using MerTK wild-type and knock-out hosts and whether inhibition of MerTK signaling with warfarin could replicate MerTK knock-out phenotypes. METHODS AND MATERIALS: Wild-type and MerTK-/- BALB/c mice were grafted in the flanks with CT26 tumors and treated with computed tomography guided RT. The role of macrophages and CD8 T cells in the response to radiation were demonstrated with cell depletion studies. The role of MerTK in priming immune responses after RT alone and with agonist antibodies to the T cell costimulatory molecule OX40 was evaluated in a Panc02-SIY model antigen system. The effect of warfarin therapy on the in-field and abscopal response to RT was demonstrated in murine models of colorectal adenocarcinoma. The association between warfarin and progression-free survival for patients treated with SABR for early-stage non-small cell lung cancer was evaluated in a multi-institutional retrospective study. RESULTS: MerTK-/- hosts had better tumor control after RT compared with wild-type mice in a macrophage and CD8 T cell-dependent manner. MerTK-/- mice showed increased counts of tumor antigen-specific CD8 T cells in the peripheral blood after tumor-directed RT alone and in combination with agonist anti-OX40. Warfarin therapy phenocopied MerTK-/- for single-flank tumors treated with RT and improved abscopal responses for RT combined with anti-CTLA4. Patients on warfarin therapy when treated with SABR for non-small cell lung cancer had higher progression-free survival rates compared with non-warfarin users. CONCLUSIONS: MerTK inhibits adaptive immune responses after SABR. Because warfarin inhibits MerTK signaling and phenocopies genetic deletion of MerTK in mice, warfarin therapy may have beneficial effects in combination with SABR and immune therapy in patients with cancer.


Subject(s)
Adaptive Immunity/genetics , Adaptive Immunity/radiation effects , Gene Knockout Techniques , c-Mer Tyrosine Kinase/deficiency , c-Mer Tyrosine Kinase/genetics , Animals , Cell Line, Tumor , Humans , Mice , Molecular Targeted Therapy , Warfarin/pharmacology , Warfarin/therapeutic use
11.
Expert Rev Gastroenterol Hepatol ; 13(11): 1039-1048, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31657973

ABSTRACT

Introduction: Environmental factors and an altered fecal microbiome are believed to be central to the pathogenesis of inflammatory bowel diseases (IBD). Vitamin D and ultraviolet radiation (UVR) are environmental factors that are associated by several pathways, including changes to the gastrointestinal microbiome, with the development and course of IBD.Area covered: This review explores the interaction of vitamin D, and UVR, with the intestinal innate and adaptive immune systems, and how they may influence the gut microbiome and the subsequent development, and progression, of IBD.Expert opinion: Vitamin D and UVR both regulate innate and adaptive immunity through a combination of common and independent mechanisms, with the overall effect being the promotion of immune tolerance. Vitamin D, and to a lesser extent UVR, can modify the gastrointestinal microbiome either directly, or through immune-mediated mechanisms and this may explain the effect on intestinal inflammation in animal models of IBD and some clinical studies. Thus, both vitamin D and UVR exposure can be considered potential 'master regulators' of gastrointestinal immunity, fine-tuning the complex interaction between genetics, host immunity and the gut microbiome. Further research and increased understanding of environment-host interactions is essential to achieving the ultimate goal of preventing and curing IBD.


Subject(s)
Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Environmental Exposure , Feces/microbiology , Gastrointestinal Microbiome , Ultraviolet Rays , Vitamin D Deficiency , Vitamin D/administration & dosage , Adaptive Immunity/drug effects , Adaptive Immunity/radiation effects , Animals , Colitis, Ulcerative/epidemiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Crohn Disease/epidemiology , Crohn Disease/immunology , Crohn Disease/microbiology , Dysbiosis , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/radiation effects , Host-Pathogen Interactions , Humans , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Prognosis , Risk Factors , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/immunology , Vitamin D Deficiency/microbiology
12.
Nat Rev Immunol ; 19(11): 688-701, 2019 11.
Article in English | MEDLINE | ID: mdl-31213673

ABSTRACT

Ultraviolet (UV) radiation is a ubiquitous component of the environment that has important effects on a wide range of cell functions. Short-wavelength UVB radiation induces sunburn and is a potent immunomodulator, yet longer-wavelength, lower-energy UVA radiation also has effects on mammalian immunity. This Review discusses current knowledge regarding the mechanisms by which UV radiation can modify innate and adaptive immune responses and how this immunomodulatory capacity can be both beneficial in the case of inflammatory and autoimmune diseases, and detrimental in the case of skin cancer and the response to several infectious agents.


Subject(s)
Immune System/radiation effects , Ultraviolet Rays , Adaptive Immunity/radiation effects , Antimicrobial Cationic Peptides/physiology , DNA Damage , Humans , Immunity, Innate , Receptors, Aryl Hydrocarbon/physiology , Receptors, Pattern Recognition/physiology , Urocanic Acid/pharmacology
13.
Br J Dermatol ; 180(3): 604-614, 2019 03.
Article in English | MEDLINE | ID: mdl-30307614

ABSTRACT

BACKGROUND: Sun protection factor (SPF) is assessed with sunscreen applied at 2 mg cm-2 . People typically apply around 0·8 mg cm-2 and use sunscreen daily for holidays. Such use results in erythema, which is a risk factor for skin cancer. OBJECTIVES: To determine (i) whether typical sunscreen use resulted in erythema, epidermal DNA damage and photoimmunosuppression during a sunny holiday, (ii) whether optimal sunscreen use inhibited erythema and (iii) whether erythema is a biomarker for photoimmunosuppression in a laboratory study. METHODS: Holidaymakers (n = 22) spent a week in Tenerife (very high ultraviolet index) using their own sunscreens without instruction (typical sunscreen use). Others (n = 40) were given SPF 15 sunscreens with instructions on how to achieve the labelled SPF (sunscreen intervention). Personal ultraviolet radiation (UVR) exposure was monitored electronically as the standard erythemal dose (SED) and erythema was quantified. Epidermal cyclobutane pyrimidine dimers (CPDs) were determined by immunostaining, and immunosuppression was assessed by contact hypersensitivity (CHS) response. RESULTS: There was no difference between personal UVR exposure in the typical sunscreen use and sunscreen intervention groups (P = 0·08). The former had daily erythema on five UVR-exposed body sites, increased CPDs (P < 0·001) and complete CHS suppression (20 of 22). In comparison, erythema was virtually absent (P < 0·001) when sunscreens were used at ≥ 2 mg cm-2 . A laboratory study showed that 3 SED from three very different spectra suppressed CHS by around ~50%. CONCLUSIONS: Optimal sunscreen use prevents erythema during a sunny holiday. Erythema predicts suppression of CHS (implying a shared action spectrum). Given that erythema and CPDs share action spectra, the data strongly suggest that optimal sunscreen use will also reduce CPD formation and UVR-induced immunosuppression.


Subject(s)
Erythema/prevention & control , Sunlight/adverse effects , Sunscreening Agents/administration & dosage , Adaptive Immunity/drug effects , Adaptive Immunity/radiation effects , Adult , DNA Damage/drug effects , DNA Damage/radiation effects , Erythema/etiology , Erythema/immunology , Female , Holidays , Humans , Immune Tolerance/drug effects , Immune Tolerance/radiation effects , Male , Middle Aged , Skin Neoplasms/etiology , Skin Neoplasms/prevention & control , Spain , Sun Protection Factor , Sunscreening Agents/chemistry
14.
J Biophotonics ; 12(2): e201800120, 2019 02.
Article in English | MEDLINE | ID: mdl-30203577

ABSTRACT

As low-level laser therapy immune cells responses are not always clarified, this study aimed to evaluate cytokines and immune cells profile after low-level laser therapy (LLLT) on arthritis-induced model. Arthritis was induced in C57BL/6 mice divided into five groups: euthanized 5 hours after inflammation induction; untreated; dexamethasone treated; LLLT at 3 Jcm-2 ; LLLT at 30 Jcm-2 . Cytokine measurements by enzyme-linked immunosorbent assay and mRNA cytokine relative levels by real-time quantitative polymerase chain reaction were performed with arthritic ankle (IL-1ß, IL-6, TNF-α, IL-10 and TGF-ß). Macrophages, dendritic cells, natural killer cells, lymphocytes CD4+ , CD8+ , Treg and costimulatory proteins were quantified in proximal lymph node by flow cytometry. Data showed decrease in all cytokine levels after LLLT and alteration in mRNA relative levels, depending on the energy density used. LLLT was able to increase of immune cell populations analyzed in the lymph node as well as costimulatory proteins expression on macrophages and dendritic cells. Treg TCD4+ and TCD8+ population enrichment were observed in LLLT at 3 and 30 Jcm-2 groups, respectively. Furthermore, Treg TCD8+ cells expressing higher levels of CD25 were observed at LLLT at 30 Jcm-2 group. Our results indicate that LLLT could change the inflammatory course of arthritis, tending to accelerate its resolution through immune cells photobiostimulation.


Subject(s)
Arthritis/immunology , Arthritis/therapy , Low-Level Light Therapy , Adaptive Immunity/radiation effects , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/radiation effects , Arthritis/metabolism , Arthritis/pathology , Cytokines/metabolism , Male , Mice , Mice, Inbred C57BL
15.
Electromagn Biol Med ; 38(1): 84-95, 2019.
Article in English | MEDLINE | ID: mdl-30518268

ABSTRACT

Extremely low frequency electromagnetic field (ELF-EMF) is produced extensively in modern technologies. Numerous in vitro and in vivo studies have shown that ELF-EMF has both stimulatory and inhibitory effects on the immune system response. This review was conducted on effects of ELF-EMF on cytokines of innate and adaptive immunity. Mechanisms of ELF-EMF, which may modulate immune cell responses, were also studied. Physical and biological parameters of ELF-EMF can interact with each other to create beneficial or harmful effect on the immune cell responses by interfering with the inflammatory or anti-inflammatory cytokines. According to the studies, it is supposed that short-term (2-24 h/d up to a week) exposure of ELF-EMF with strong density may increase innate immune response due to an increase of innate immunity cytokines. Furthermore, long-term (2-24 h/d up to 8 years) exposure to low-density ELF-EMF may cause a decrease in adaptive immune response, especially in Th1 subset.


Subject(s)
Adaptive Immunity/radiation effects , Electromagnetic Fields , Immunity, Innate/radiation effects , Animals , Cytokines/metabolism , Humans
16.
EMBO Mol Med ; 10(12)2018 12.
Article in English | MEDLINE | ID: mdl-30442705

ABSTRACT

Emerging evidence suggests a role for radiation in eliciting anti-tumour immunity. We aimed to investigate the role of macrophages in modulating the immune response to radiation. Irradiation to murine tumours generated from colorectal (MC38) and pancreatic (KPC) cell lines induced colony-stimulating factor 1 (CSF-1). Coincident with the elevation in CSF-1, macrophages increased in tumours, peaking 5 days following irradiation. These tumour-associated macrophages (TAMs) were skewed towards an immunosuppressive phenotype. Macrophage depletion via anti-CSF (aCSF) reduced macrophage numbers, yet only achieved tumour growth delay when combined with radiation. The tumour growth delay from aCSF after radiation was abrogated by depletion of CD8 T cells. There was enhanced recognition of tumour cell antigens by T cells isolated from irradiated tumours, consistent with increased antigen priming. The addition of anti-PD-L1 (aPD-L1) resulted in improved tumour suppression and even regression in some tumours. In summary, we show that adaptive immunity induced by radiation is limited by the recruitment of highly immunosuppressive macrophages. Macrophage depletion partly reduced immunosuppression, but additional treatment with anti-PD-L1 was required to achieve tumour regression.


Subject(s)
Adaptive Immunity/radiation effects , Colorectal Neoplasms/radiotherapy , Leukocyte Reduction Procedures , Macrophages/immunology , Pancreatic Neoplasms/radiotherapy , X-Ray Therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Disease Models, Animal , Mice , Pancreatic Neoplasms/immunology , Treatment Outcome
17.
Int J Mol Sci ; 19(11)2018 Nov 13.
Article in English | MEDLINE | ID: mdl-30428512

ABSTRACT

In cancer treatments, especially high-dose radiotherapy (HDRT) is applied. Patients suffering from chronic inflammatory diseases benefit from low-dose radiation therapy (LDRT), but exposure to very low radiation doses can still steadily increase for diagnostic purposes. Yet, little is known about how radiation impacts on forms of cell death in human immune cells. In this study, the radiosensitivity of human immune cells of the peripheral blood was examined in a dose range from 0.01 to 60 Gy with regard to induction of apoptosis, primary necrosis, and secondary necrosis. Results showed that immune cells differed in their radiosensitivity, with monocytes being the most radioresistant. T cells mainly died by necrosis and were moderately radiosensitive. This was followed by B and natural killer (NK) cells, which died mainly by apoptosis. X-radiation had no impact on cell death in immune cells at very low doses (≤0.1 Gy). Radiation doses of LDRT (0.3⁻0.7 Gy) impacted on the more radiosensitive NK and B cells, which might contribute to attenuation of inflammation. Even single doses applied during RT of tumors did not erase the immune cells completely. These in vitro studies can be considered as the basis to optimize individual radiation therapy schemes in multimodal settings and to define suited time points for further inclusion of immunotherapies.


Subject(s)
Adaptive Immunity/radiation effects , Immunity, Innate/radiation effects , Apoptosis/radiation effects , B-Lymphocytes/radiation effects , Cell Death/radiation effects , Dose-Response Relationship, Radiation , Humans , Killer Cells, Natural/radiation effects , Monocytes/radiation effects , Radiation Exposure/adverse effects , Radiotherapy
18.
Immunology ; 154(3): 510-521, 2018 07.
Article in English | MEDLINE | ID: mdl-29377107

ABSTRACT

Ultraviolet radiation (UVr) promotes several well-known molecular changes, which may ultimately impact on health. Some of these effects are detrimental, like inflammation, carcinogenesis and immunosuppression. On the other hand, UVr also promotes vitamin D synthesis and other beneficial effects. We recently demonstrated that exposure to very low doses of UVr on four consecutive days [repetitive low UVd (rlUVd)] does not promote an inflammatory state, nor the recruitment of neutrophils or lymphocytes, as the exposure to a single high UV dose (shUVd) does. Moreover, rlUVd reinforce the epithelium by increasing antimicrobial peptides transcription and epidermal thickness. The aim of this study was to evaluate the adaptive immune response after shUVd and rlUVd, determining T-cell and B-cell responses. Finally, we challenged animals exposed to both irradiation procedures with Staphylococcus aureus to study the overall effects of both innate and adaptive immunity during a cutaneous infection. We observed, as expected, a marked suppression of T-cell and B-cell responses after exposure to an shUVd but a novel and significant increase in both specific responses after exposure to rlUVd. However, the control of the cutaneous S. aureus infection was defective in this last group, suggesting that responses against pathogens cannot be ruled out from isolated stimuli.


Subject(s)
Adaptive Immunity/radiation effects , Radiation Exposure , Ultraviolet Rays , Animals , Antibody Formation/immunology , Antibody Formation/radiation effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Biomarkers , Cytokines/metabolism , Dermatitis/immunology , Dermatitis/metabolism , Dermatitis/microbiology , Dermatitis/prevention & control , Disease Models, Animal , Immunization , Immunophenotyping , Male , Mice , Radiation Dosage , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/prevention & control , Staphylococcus aureus/immunology , Staphylococcus aureus/radiation effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/radiation effects , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology
19.
Front Immunol ; 9: 3077, 2018.
Article in English | MEDLINE | ID: mdl-30692991

ABSTRACT

Radiation continues to play a major role in the treatment of almost every cancer type. Traditional radiation studies focused on its ability to damage DNA, but recent evidence has demonstrated that a key mechanism driving the efficacy of radiation in vivo is the immune response triggered in irradiated tissue. Innate immune cells including macrophages, dendritic cells, and natural killer cells are key mediators of the radiation-induced immune response. They regulate the sensing of radiation-mediated damage and subsequent radiation-induced inflammation. Given the importance of innate immune cells as determinants of the post-radiation anti-tumor immune response, much research has been devoted to identify ways to both enhance the innate immune response and prevent their ability to suppress ongoing immune responses. In this review, we will discuss how the innate immune system shapes anti-tumor immunity following radiation and highlight key strategies directed at the innate immune response to enhance the efficacy of radiation.


Subject(s)
Adaptive Immunity/drug effects , Antineoplastic Agents, Immunological/therapeutic use , Immunity, Innate/drug effects , Neoplasms/therapy , Tumor Microenvironment/radiation effects , Adaptive Immunity/radiation effects , Antineoplastic Agents, Immunological/pharmacology , Chemoradiotherapy/methods , Clinical Trials as Topic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/radiation effects , Humans , Immunity, Innate/radiation effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/radiation effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/radiation effects , Molecular Targeted Therapy/methods , Neoplasms/immunology , Treatment Outcome , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
20.
Nat Rev Clin Oncol ; 14(6): 365-379, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28094262

ABSTRACT

Investigations into the interaction between radiotherapy and the host immune system have uncovered new mechanisms that can potentially be exploited to improve the efficacy of radiotherapy. Radiation promotes the release of danger signals and chemokines that recruit inflammatory cells into the tumour microenvironment, including antigen-presenting cells that activate cytotoxic T-cell function. By contrast, radiation can attract immunosuppressive cells into the tumour microenvironment. In rare circumstances, the antitumour effect of radiotherapy has been observed outside of the radiation field, known as the abscopal effect. This phenomenon is proposed to have an immune origin and indicates that local radiotherapy elicits systemic effects. Herein, we highlight data that provide new mechanistic explanations for the success or failure of radiotherapy, and postulate how the combination of immune-modulation and radiation could tip the balance of the host immune response to promote cure. We use the concept of radiation- induced tumour equilibrium (RITE) as a starting point to discuss the mechanistic influence of immune-checkpoint therapies on radiotherapy efficacy.


Subject(s)
Immunotherapy , Neoplasms/therapy , Adaptive Immunity/radiation effects , Combined Modality Therapy , Humans , Immunity, Innate/radiation effects , Neoplasms/immunology , Neoplasms/radiotherapy , Tumor Microenvironment/immunology , Tumor Microenvironment/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...