Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Cancer Lett ; 523: 57-71, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34563641

ABSTRACT

High fluence low-level laser (HF-LLL), a mitochondria-targeted tumour phototherapy, results in oxidative damage and apoptosis of tumour cells, as well as damage to normal tissue. To circumvent this, the therapeutic effect of low fluence LLL (LFL), a non-invasive and drug-free therapeutic strategy, was identified for tumours and the underlying molecular mechanisms were investigated. We observed that LFL enhanced antigen-specific immune response of macrophages and dendritic cells by upregulating MHC class II, which was induced by mitochondrial reactive oxygen species (ROS)-activated signalling, suppressing tumour growth in both CD11c-DTR and C57BL/6 mice. Mechanistically, LFL upregulated MHC class II in an MHC class II transactivator (CIITA)-dependent manner. LFL-activated protein kinase C (PKC) promoted the nuclear translocation of CIITA, as inhibition of PKC attenuated the DNA-binding efficiency of CIITA to MHC class II promoter. CIITA mRNA and protein expression also improved after LFL treatment, characterised by direct binding of Src and STAT1, and subsequent activation of STAT1. Notably, scavenging of ROS downregulated LFL-induced Src and PKC activation and antagonised the effects of LFL treatment. Thus, LFL treatment altered the adaptive immune response via the mitochondrial ROS-activated signalling pathway to control the progress of neoplastic disease.


Subject(s)
Histocompatibility Antigens Class II/immunology , Low-Level Light Therapy/methods , Neoplasms, Experimental/therapy , Protein Kinase C/physiology , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , src-Family Kinases/physiology , Active Transport, Cell Nucleus , Animals , Antigen Presentation , Dendritic Cells/physiology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Nuclear Proteins/physiology , STAT1 Transcription Factor/physiology , Trans-Activators/physiology
2.
Immunobiology ; 225(2): 151896, 2020 03.
Article in English | MEDLINE | ID: mdl-31870642

ABSTRACT

We recently reported identification of sarcoplasmic/endoplasmic reticulum calcium-ATPase2a (SERCA2a) 971-990, which induces atrial myocarditis by generating autoreactive T cells in A/J mice. However, it was unknown how antigen-sensitized T cells could recognize SERCA2a 971-990, since SERCA2a-expression is confined to an intracellular compartment. In this report, we present evidence that antigen-presenting cells (APCs) from lymphoid and non-lymphoid organs in naïve animals present SERCA2a 971-990 and stimulate antigen-specific T cells. Using major histocompatibility complex (MHC) class II dextramers for SERCA2a 971-990, we created a panel of T cell hybridomas and demonstrated that splenocytes from naïve A/J mice stimulated the hybridoma cells without exogenous supplementation of SERCA2a 971-990. We then recapitulated this phenomenon by using SERCA2a 971-990 -specific primary T cells, verifying that the T cell responses were MHC-restricted. Furthermore, SERCA2a 971-990 -sensitzed T cells exposed to APCs from naïve mice were found to produce the inflammatory cytokines interferon-γ, granulocyte macrophage colony stimulating factor, and interleukin-17A, which are implicated in the induction of myocarditis. Finally, while T cells exposed to mononuclear cells (MNCs) obtained from heart and liver also responded similarly to splenocytes, endothelial cells (ECs) generated from the corresponding organs displayed opposing effects, in that the proliferative responses were suppressed with the heart ECs, but not with the liver ECs. Taken together, our data suggest that the surface expression of SERCA2a 971-990 by naïve APCs can potentially trigger pathogenic autoreactive T cell responses under conditions of autoimmunity, which may have implications in endothelial dysfunction.


Subject(s)
Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , Autoimmunity/immunology , Epitopes/immunology , Myocarditis/immunology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology , Animals , Cytokines/immunology , Endothelial Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Histocompatibility Antigens Class II/immunology , Hybridomas/immunology , Inflammation/immunology , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred Strains , T-Lymphocytes
3.
ACS Infect Dis ; 5(11): 1867-1878, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31498993

ABSTRACT

Lipid-based nanoparticles have in recent years attracted increasing attention as pharmaceutical carriers. In particular, reports of them having inherent adjuvant properties combined with their ability to protect antigen from degradation make them suitable as vaccine vectors. However, the physicochemical profile of an ideal nanoparticle for vaccine delivery is still poorly defined. Here, we used an in vitro dendritic cell assay to assess the immunogenicity of a variety of liposome formulations as vaccine carriers and adjuvants. Using flow cytometry, we investigated liposome-assisted antigen presentation as well as the expression of relevant costimulatory molecules on the cell surface. Cytokine secretion was further evaluated with an enzyme-linked immunosorbent assay (ELISA). We show that liposomes can successfully enhance antigen presentation and maturation of dendritic cells, as compared to vaccine fusion protein (CTA1-3Eα-DD) administered alone. In particular, the lipid phase state of the membrane was found to greatly influence the vaccine antigen processing by dendritic cells. As compared to their fluid phase counterparts, gel phase liposomes were more efficient at improving antigen presentation. They were also superior at upregulating the costimulatory molecules CD80 and CD86 as well as increasing the release of the cytokines IL-6 and IL-1ß. Taken together, we demonstrate that gel phase liposomes, while nonimmunogenic on their own, significantly enhance the antigen-presenting ability of dendritic cells and appear to be a promising way forward to improve vaccine immunogenicity.


Subject(s)
Dendritic Cells/immunology , Histocompatibility Antigens Class II/immunology , Liposomes/immunology , Phosphatidylcholines/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Antigen Presentation , B7-1 Antigen/immunology , Cells, Cultured , Cytokines/immunology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Female , Liposomes/chemistry , Mice , Mice, Inbred C57BL , Phosphatidylcholines/immunology , Vaccines/chemistry , Vaccines/pharmacology
4.
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
5.
Circulation ; 138(11): 1130-1143, 2018 09 11.
Article in English | MEDLINE | ID: mdl-29588316

ABSTRACT

BACKGROUND: CD4+ T cells play an important role in atherosclerosis, but their antigen specificity is poorly understood. Immunization with apolipoprotein B (ApoB, core protein of low density lipoprotein) is known to be atheroprotective in animal models. Here, we report on a human APOB peptide, p18, that is sequence-identical in mouse ApoB and binds to both mouse and human major histocompatibility complex class II molecules. METHODS: We constructed p18 tetramers to detect human and mouse APOB-specific T cells and assayed their phenotype by flow cytometry including CD4 lineage transcription factors, intracellular cytokines, and T cell receptor activation. Apolipoprotein E-deficient ( Apoe-/-) mice were vaccinated with p18 peptide or adjuvants alone, and atherosclerotic burden in the aorta was determined. RESULTS: In human peripheral blood mononuclear cells from donors without cardiovascular disease, p18 specific CD4+ T cells detected by a new human leukocyte antigen-antigen D related-p18 tetramers were mostly Foxp3+ regulatory T cells (Tregs). Donors with subclinical cardiovascular disease as detected by carotid artery ultrasound had Tregs coexpressing retinoic acid-related orphan receptor gamma t or T-bet, which were both almost absent in donors without cardiovascular disease. In Apoe-/- mice, immunization with p18 induced Tregs and reduced atherosclerotic lesions. After peptide restimulation, responding CD4+ T cells identified by Nur77-GFP (green fluorescent protein) were highly enriched in Tregs. A new mouse I-Ab-p18 tetramer identified the expansion of p18-specific CD4+ T cells on vaccination, which were enriched for interleukin-10-producing Tregs. CONCLUSIONS: These findings show that APOB p18-specific CD4+ T cells are mainly Tregs in healthy donors, but coexpress other CD4 lineage transcription factors in donors with subclinical cardiovascular disease. This study identifies ApoB peptide 18 as the first Treg epitope in human and mouse atherosclerosis.


Subject(s)
Apolipoprotein B-100/immunology , Apolipoproteins B/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Peptide Fragments/immunology , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Aorta/immunology , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Disease Models, Animal , Epitope Mapping , Female , Freund's Adjuvant/administration & dosage , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Peptide Fragments/administration & dosage , Plaque, Atherosclerotic , Vaccination
6.
Scand J Immunol ; 87(3)2018 Mar.
Article in English | MEDLINE | ID: mdl-29412476

ABSTRACT

Taking into consideration the multiparametric nature of systemic lupus erythematosus (SLE), the severity and variability of symptoms and the lack of effective therapeutic approaches, this study took advantage of the recently described role of soluble major histocompatibility complex class II (sMHCII) molecules in maintaining tolerance to the organism and attempted to apply sMHCII proteins as a treatment to murine SLE experimental models in vitro as well as in vivo. After breaking tolerance to DNA in vitro, which was accompanied by development of specific anti-dsDNA antibodies, syngeneic or allogeneic sMHCII molecules, purified from healthy mouse serum, could significantly reduce the specific antibody levels and drive the system towards immunosuppression, as assessed by specific marker analysis on T cells and cytokine production by flow cytometry and ELISA, respectively. The in vivo experimental model consisted of pristane-induced SLE symptoms to BALB/c mice, which developed maximal levels of anti-dsDNA 2 months after pristane inoculation. Syngeneic or allogeneic sMHCII administration could alleviate pristane-induced symptoms, significantly decrease specific anti-dsDNA antibody production and develop immunosuppression to the host, as manifested by increase of CD4 + CTLA-4 +  and CD4 + CD25 +  cell populations in the spleen. Thus, the results presented in this study introduced the ability of sMHCII proteins to suppress specific autoantigen response, opening new areas of research and offering novel therapeutic approaches to SLE with expanding features to other autoimmune diseases.


Subject(s)
Antibodies, Antinuclear/immunology , Autoantigens/immunology , Histocompatibility Antigens Class II/immunology , Immune Tolerance/immunology , Immunotherapy/methods , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/therapy , T-Lymphocytes/immunology , Animals , CD4 Antigens/metabolism , CTLA-4 Antigen/metabolism , Cells, Cultured , DNA/immunology , Disease Models, Animal , Immunosuppression Therapy , Interleukin-2 Receptor alpha Subunit/metabolism , Lupus Erythematosus, Systemic/chemically induced , Mice , Mice, Inbred BALB C , Spleen/cytology , Spleen/immunology , Terpenes/adverse effects
7.
Mol Immunol ; 93: 133-143, 2018 01.
Article in English | MEDLINE | ID: mdl-29175593

ABSTRACT

Human schistosomiasis is a neglected tropical disease of great importance in public health. A large number of people are infected with schistosomiasis, making vaccine development and effective diagnosis important control strategies. A rational epitope prediction workflow using Schistosoma mansoni hypothetical proteins was previously presented by our group, and an improvement to that approach is presented here. Briefly, immunodominant epitopes from parasite membrane proteins were predicted by reverse vaccinology strategy with additional in silico analysis. Furthermore, epitope recognition was evaluated using sera of individuals infected with S. mansoni. The epitope that stood out in both in silico and in vitro assays was used to compose a rational chimeric molecule to improve immune response activation. Out of 2185 transmembrane proteins, four epitopes with high binding affinities for human and mouse MHCII molecules were selected through computational screening. These epitopes were synthesized to evaluate their ability to induce TCD4+ lymphocyte proliferation in mice. Sm204830e and Sm043300e induced significant TCD4+ proliferation. Both epitopes were submitted to enzyme-linked immunosorbent assay to evaluate their recognition by IgG antibodies from the sera of infected individuals, and epitope Sm043300 was significantly recognized in most sera samples. Epitope Sm043300 also showed good affinity for human MHCII molecules in molecular docking, and its sequence is curiously highly conserved in four S. mansoni proteins, all of which are described as G-protein-coupled receptors. In addition, we have demonstrated the feasibility of incorporating this epitope, which showed low similarity to human sequences, into a chimeric molecule. The stability of the molecule was evaluated by molecular modeling aimed at future molecule production for use in diagnosis and vaccination trials.


Subject(s)
Antigens, Helminth/immunology , Immunodominant Epitopes/immunology , Schistosoma mansoni/immunology , Amino Acid Sequence , Animals , Antibodies, Helminth/blood , Antibodies, Helminth/immunology , Antigens, Helminth/genetics , CD4-Positive T-Lymphocytes/immunology , Combinatorial Chemistry Techniques , Drug Design , Drug Evaluation, Preclinical , Female , HLA-DRB1 Chains/immunology , Helminth Proteins/chemistry , Helminth Proteins/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunodominant Epitopes/genetics , Immunodominant Epitopes/metabolism , Lymphocyte Activation , Membrane Proteins/chemistry , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Docking Simulation , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Schistosoma haematobium/immunology , Schistosoma mansoni/genetics , Schistosomiasis mansoni/blood , Schistosomiasis mansoni/immunology , Sequence Alignment , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
8.
Vaccine ; 35(7): 1037-1045, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28111143

ABSTRACT

Activating innate immunity by an adjuvant is required in vaccine development. The study aims to investigate adjuvant effects of aqueous extracts of Artemisia rupestris L. (AEAR) in vivo and in vitro. ICR mice were subcutaneously administered with antigen and AEAR at various doses to evaluate their immune responses of antibodies, dendritic cells (DCs), regulatory T cells (Treg), splenic lymphocyte, and cytokine. The evaluation results showed that AEAR could largely increase titers of antigen-specific antibodies (IgG, IgG1, and IgG2a) and T cell proliferation. AEAR also increased expression of IFN-γ in CD8+T cells as well as IL-4 and INF-γ expression in CD4+T cells. Expression levels of MHC-II, CD40, CD80, and CD86 on DCs were significantly elevated, whereas the Treg frequency was significantly decreased. AEAR (200µg) showed remarkable adjuvant activity. Furthermore, AEAR enhanced MHC-II, CD40, CD80, and CD86 expression as well as the yields of TNF-α and IL-12 on DCs through toll-like receptor4 (TLR4) in vitro. Those results indicated that AEAR could serve as an efficacious immune stimulator for vaccines because it significantly enhanced specific immune responses by promoting DCs maturation and reduced Treg through TLR4 signaling pathway.


Subject(s)
Adjuvants, Immunologic/pharmacology , Artemisia/chemistry , Ovalbumin/immunology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , Toll-Like Receptor 4/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Gene Expression Regulation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Immunoglobulin G/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12/genetics , Interleukin-12/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphocyte Activation , Mice , Mice, Inbred ICR , Ovalbumin/administration & dosage , Plant Extracts/chemistry , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Transplantation ; 100(8): 1683-91, 2016 08.
Article in English | MEDLINE | ID: mdl-27362308

ABSTRACT

BACKGROUND: The dual role of B cells as drivers and suppressors of the immune responses have underscored the need to trace the fate of B cells recognizing donor major histocompatibility complex class I and class II after allograft transplantation. METHODS: In this study, we used donor class II tetramers to trace the fate of I-E-specific B cells after immunization with BALB/c spleen cells or cardiac transplantation, in naive or sensitized C57BL/6 recipients. We combined this approach with genetic lineage tracing of memory B cells in activation-induced cytidine deaminase regulated Cre transgenic mice crossed to the ROSA26-enhanced yellow fluorescent protein reporter mice to track endogenous I-E-specific memory B cell generation. RESULTS: Immunization with BALB/c splenocytes or heart transplantation induced an expansion and differentiation of I-E-specific B cells into germinal center B cells, whereas BALB/c heart transplantation into sensitized recipients induced the preferential differentiation into antibody-secreting cells. A 10.8-fold increase in the frequency of I-E-specific memory B cells was observed by day 42 postimmunization. Treatment with CTLA4-Ig starting on day 0 or day 7 postimmunization abrogated I-E-specific memory B cell generation and sensitized humoral responses, but not if treatment commenced on day 14. CONCLUSIONS: The majority of donor-specific memory B cells are generated between days 7 and 14 postimmunization, thus revealing a flexible timeframe whereby delayed CTLA4-Ig administration can inhibit sensitization and the generation of memory graft-reactive B cells.


Subject(s)
Abatacept/administration & dosage , B-Lymphocytes/drug effects , Cell Lineage , Cell Proliferation/drug effects , Graft Rejection/prevention & control , Heart Transplantation/adverse effects , Histocompatibility Antigens Class II/immunology , Immunologic Memory , Immunosuppressive Agents/administration & dosage , Lymphocyte Activation/drug effects , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Cell Tracking/methods , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Disease Models, Animal , Drug Administration Schedule , Genotype , Graft Rejection/blood , Graft Rejection/genetics , Graft Rejection/immunology , Graft Survival/drug effects , Histocompatibility Antigens Class II/blood , Integrases/genetics , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , RNA, Untranslated/genetics , Time Factors
10.
Pharm Biol ; 54(7): 1125-32, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26067828

ABSTRACT

CONTEXT: Thyme has been used in traditional medicine for medicinal purposes since ancient times. OBJECTIVE: The objective of this study was to investigate the effects of thymol and carvacrol as two major constituents of thyme on dendritic cells (DCs) maturation and T cell activation. MATERIALS AND METHODS: Splenic DCs were treated with non-cytotoxic concentrations of the components and then analyzed for MHC II, CD86, and CD40 expression by flow cytometry. The effects of compounds on mitogenic, as well as allogenic T cell responses in mixed lymphocyte culture (MLR) and the release of cytokines were investigated. RESULTS: At 0.1 µg/ml, reduced mean fluorescent intensity (MFI) of CD86 for thymol (80.3 ± 0.2% of untreated control) and CD40 for carvacrol (79.5 ± 0.14%) was observed (p < 0.001). Decreased mitogenic T cell proliferation by thymol [proliferation index (PI) from 0.93 ± 0.11 at 1 µg/ml to 0.42 ± 0.16 at 100 µg/ml (p < 0.01)] and carvacrol [PI from 1.08 ± 0.3 at 1 µg/ml to 0.28 ± 0.1 at 100 µg/ml (p < 0.001)] was seen. Ten micrograms/ml thymol (PI, 0.85 ± 0.04) and carvacrol (PI, 0.89 ± 0.03) inhibited allogenic T cell response (p < 0.05). Decreased IFN-γ level in MLR supernatant from 1441 ± 27.7 pg/ml in untreated cells to 944 ± 32.1 at 10 µg/ml of thymol and of carvacrol (886 ± 31.7 pg/ml) (p < 0.01) was found. IL-4 levels were decreased in the presence of both compounds (p < 0.01). CONCLUSION: These data showed the suppressive effects of thymol and carvacrol on DCs maturation and function, as well as T cell responses.


Subject(s)
Dendritic Cells/drug effects , Immunosuppressive Agents/pharmacology , Monoterpenes/pharmacology , Thymol/pharmacology , Thymus Plant , Animals , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD40 Antigens/immunology , CD40 Antigens/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Cymenes , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dose-Response Relationship, Drug , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immunosuppressive Agents/isolation & purification , Lymphocyte Activation/drug effects , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Monoterpenes/isolation & purification , Phenotype , Phytotherapy , Plants, Medicinal , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymol/isolation & purification , Thymus Plant/chemistry
11.
Cell Death Dis ; 6: e1761, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25973681

ABSTRACT

One prerequisite that radiotherapy (RT) and chemotherapy (CT) result in anti-tumor immune responses is triggering of immunogenic cell death forms such as necroptosis. The latter is inducible by inhibition of apoptosis with the pan-caspase inhibitor zVAD-fmk. The design of multimodal therapies that overcome melanoma's resistance to apoptosis is a big challenge of oncoimmunology. As hints exist that immune stimulation by hyperthermia (HT) augments the efficacy of melanoma therapies and that tumors can be sensitized for RT with zVAD-fmk, we asked whether combinations of RT with dacarbazine (DTIC) and/or HT induce immunogenic melanoma cell death and how this is especially influenced by zVAD-fmk. Necroptosis was inducible in poorly immunogenic B16-F10 melanoma cells and zVAD-fmk generally increased melanoma cell necrosis concomitantly with the release of HMGB1. Supernatants (SNs) of melanoma cells whose cell death was modulated with zVAD-fmk induced an upregulation of the activation markers CD86 and MHCII on macrophages. The same was seen on dendritic cells (DCs), but only when zVAD-fmk was added to multimodal tumor treatments including DTIC. DCs of MyD88 KO mice and DCs incubated with SNs containing apyrase did not increase the expression of these activation markers on their surface. The in vivo experiments revealed that zVAD-fmk decreases the tumor growth significantly and results in a significantly reduced tumor infiltration of Tregs when added to multimodal treatment of the tumor with RT, DTIC and HT. Further, a significantly increased DC and CD8+ T-cell infiltration into the tumor and in the draining lymph nodes was induced, as well as an increased expression of IFNγ by CD8+ T cells. However, zVAD-fmk did not further reduce tumor growth in MyD88 KO mice, mice treated with apyrase or RAG KO mice. We conclude that HMGB1, nucleotides and CD8+ T cells mediate zVAD-fmk induced anti-melanoma immune reactions in multimodal therapy settings.


Subject(s)
Amino Acid Chloromethyl Ketones/therapeutic use , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , HMGB1 Protein/metabolism , Melanoma, Experimental/pathology , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Apyrase/therapeutic use , B7-2 Antigen/biosynthesis , B7-2 Antigen/immunology , Caspase Inhibitors/therapeutic use , Cell Line, Tumor , Chemoradiotherapy , Combined Modality Therapy , Dacarbazine/therapeutic use , Dendritic Cells/immunology , Histocompatibility Antigens Class II/biosynthesis , Histocompatibility Antigens Class II/immunology , Homeodomain Proteins/genetics , Hyperthermia, Induced , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Macrophages, Peritoneal/immunology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Radiation, Ionizing , Tumor Necrosis Factor-alpha/metabolism
12.
Clin Exp Allergy ; 45(6): 1015-26, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25900315

ABSTRACT

Allergen immunotherapy (AIT) has been practised since 1911 and remains the only therapy proven to modify the natural history of allergic diseases. Although efficacious in carefully selected individuals, the currently licensed whole allergen extracts retain the risk of IgE-mediated adverse events, including anaphylaxis and occasionally death. This together with the need for prolonged treatment regimens results in poor patient adherence. The central role of the T cell in orchestrating the immune response to allergen informs the choice of T cell targeted therapies for down-regulation of aberrant allergic responses. Carefully mapped short synthetic peptides that contain the dominant T cell epitopes of major allergens and bind to a diverse array of HLA class II alleles, can be delivered intradermally into non-inflamed skin to induce sustained clinical and immunological tolerance. The short peptides from allergenic proteins are unable to cross-link IgE and possess minimal inflammatory potential. Systematic progress has been made from in vitro human models of allergen T cell epitope-based peptide anergy in the early 1990s, through proof-of-concept murine allergy models and early human trials with longer peptides, to the current randomized, double-blind, placebo-controlled clinical trials with the potential new class of synthetic short immune-regulatory T cell epitope peptide therapies. Sustained efficacy with few adverse events is being reported for cat, house dust mite and grass pollen allergy after only a short course of treatment. Underlying immunological mechanisms remain to be fully delineated but anergy, deletion, immune deviation and Treg induction all seem contributory to successful outcomes, with changes in IgG4 apparently less important compared to conventional AIT. T cell epitope peptide therapy is promising a safe and effective new class of specific treatment for allergy, enabling wider application even for more severe allergic diseases.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunomodulation , Peptides/immunology , Allergens/chemistry , Allergens/immunology , Animals , Antigen Presentation/immunology , Clinical Trials as Topic , Disease Models, Animal , Drug Evaluation, Preclinical , Epitope Mapping/methods , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/therapeutic use , Histocompatibility Antigens Class II/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunologic Factors/therapeutic use , Immunotherapy , Molecular Targeted Therapy , Peptides/chemistry , Peptides/therapeutic use , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Translational Research, Biomedical , Treatment Outcome
13.
Proc Natl Acad Sci U S A ; 112(19): 6146-51, 2015 May 12.
Article in English | MEDLINE | ID: mdl-25902531

ABSTRACT

At their margins, tumors often contain neutrophils, dendritic cells, and activated macrophages, which express class II MHC and CD11b products. The interplay between stromal cells, tumor cells, and migratory cells such as lymphocytes creates opportunities for noninvasive imaging of immune responses. We developed alpaca-derived antibody fragments specific for mouse class II MHC and CD11b products, expressed on the surface of a variety of myeloid cells. We validated these reagents by flow cytometry and two-photon microscopy to obtain images at cellular resolution. To enable noninvasive imaging of the targeted cell populations, we developed a method to site-specifically label VHHs [the variable domain (VH) of a camelid heavy-chain only antibody] with (18)F or (64)Cu. Radiolabeled VHHs rapidly cleared the circulation (t1/2 ≈ 20 min) and clearly visualized lymphoid organs. We used VHHs to explore the possibility of imaging inflammation in both xenogeneic and syngeneic tumor models, which resulted in detection of tumors with remarkable specificity. We also imaged the infiltration of myeloid cells upon injection of complete Freund's adjuvant. Both anti-class II MHC and anti-CD11b VHHs detected inflammation with excellent specificity. Given the ease of manufacture and labeling of VHHs, we believe that this method could transform the manner in which antitumor responses and/or infectious events may be tracked.


Subject(s)
Immune System/physiology , Neoplasms/immunology , Positron-Emission Tomography , Aminoacyltransferases/physiology , Animals , Antibodies/immunology , Antineoplastic Agents/therapeutic use , Bacterial Proteins/physiology , Bone Marrow Cells/metabolism , Copper Radioisotopes/chemistry , Cysteine Endopeptidases/physiology , Flow Cytometry , Fluorine Radioisotopes/chemistry , Freund's Adjuvant , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Inflammation , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Neoplasm Transplantation , Neoplasms/therapy
14.
Arthritis Rheumatol ; 67(8): 2061-70, 2015 May.
Article in English | MEDLINE | ID: mdl-25892196

ABSTRACT

OBJECTIVE: The mechanisms underlying bone damage in rheumatoid arthritis (RA) are incompletely understood. We recently identified the shared epitope (SE), an HLA-DRB1-coded 5-amino acid sequence motif carried by the majority of RA patients as a signal transduction ligand that interacts with cell surface calreticulin and accelerates osteoclast (OC)-mediated bone damage in collagen-induced arthritis (CIA). Given the role of the SE/calreticulin pathway in arthritis-associated bone damage, we sought to determine the therapeutic targetability of calreticulin. METHODS: A library of backbone-cyclized peptidomimetic compounds, all carrying an identical core DKCLA sequence, was synthesized. The ability of these compounds to inhibit SE-activated signaling and OC differentiation was tested in vitro. The effect on disease severity and OC-mediated bone damage was studied by weekly intraperitoneal administration of the compounds to DBA/1 mice with CIA. RESULTS: Two members of the peptidomimetics library were found to have SE-antagonistic effects and antiosteoclast differentiation effects at picomolar concentrations in vitro. The lead mimetic compound, designated HS(4-4)c Trp, potently ameliorated arthritis and bone damage in vivo when administered in picogram doses to mice with CIA. Another mimetic analog, designated HS(3-4)c Trp, was found to lack activity, both in vitro and in vivo. The differential activity of the 2 analogs depended on minor differences in their respective ring sizes and correlated with distinctive geometry when computationally docked to the SE binding site on calreticulin. CONCLUSION: These findings identify calreticulin as a novel therapeutic target in erosive arthritis and provide sound rationale and early structure/activity relationships for future drug design.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Bone and Bones/drug effects , Calreticulin/drug effects , Histocompatibility Antigens Class II/drug effects , Osteoclasts/drug effects , Peptides/pharmacology , Animals , Arthritis, Experimental/immunology , Arthritis, Rheumatoid/immunology , Bone and Bones/metabolism , Calreticulin/metabolism , Cell Differentiation/drug effects , Disease Models, Animal , Epitopes , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , In Vitro Techniques , Ligands , Mice , Molecular Docking Simulation , Osteoclasts/metabolism , Peptide Library , Signal Transduction/drug effects
15.
Immunol Res ; 62(1): 3-15, 2015 May.
Article in English | MEDLINE | ID: mdl-25716614

ABSTRACT

Identification of potential epitopes that might activate the immune system has been facilitated by the employment of algorithms that use experimental data as templates. However, in order to prove the affinity and the map of interactions between the receptor (major histocompatibility complex, MHC, or T-cell receptor) and the potential epitope, further computational studies are required. Docking and molecular dynamics (MDs) simulations have been an effective source of generating structural information at molecular level in immunology. Herein, in order to provide a detailed understanding of the origins of epitope recognition and to select the best peptide candidate to develop an epitope-based vaccine, docking and MDs simulations in combination with MMGBSA free energy calculations and per-residue free energy decomposition were performed, taking as starting complexes those formed between four designed epitopes (P1-P4) from hemagglutinin (HA) of the H1N1 influenza virus and MHC-II anchored in POPC membrane. Our results revealed that the energetic contributions of individual amino acids within the pMHC-II complexes are mainly dictated by van der Waals interactions and the nonpolar part of solvation energy, whereas the electrostatic interactions corresponding to hydrogen bonds and salt bridges determine the binding specificity, being the most favorable interactions formed between p4 and MHC-II. Then, P1-P4 epitopes were synthesized and tested experimentally to compare theoretical and experimental results. Experimental results show that P4 elicited the highest strong humoral immune response to HA of the H1N1 and may induce antibodies that are cross-reactive to other influenza subtypes, suggesting that it could be a good candidate for the development of a peptide-based vaccine.


Subject(s)
Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Peptides/immunology , Animals , Epitopes/administration & dosage , Epitopes/chemistry , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Immunoglobulin G/blood , Influenza Vaccines/administration & dosage , Lipid Bilayers/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptides/administration & dosage , Peptides/chemistry , Phosphatidylcholines/chemistry , Rabbits
16.
Allergy ; 70(1): 49-58, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25236500

ABSTRACT

BACKGROUND: Fluorescence-labeled MHC class II/peptide tetramer complexes are considered as optimal tools to characterize allergen-specific CD4(+) T cells, but this technique is restricted to frequently expressed HLA class II molecules and knowledge of immunodominant epitopes. In contrast, allergen-stimulated proliferation assessed by CFSE dilution is less sophisticated and widely applicable. The major mugwort allergen, Art v 1, contains only one single, immunodominant, HLA-DR1-restricted epitope (Art v 125-36 ). Thus, essentially all Art v 1-reactive cells should be identified by a HLA-DRB1*01:01/Art v 119-36 tetramer. METHODS: We compared specificity and sensitivity of tetramer(+) and allergen-induced proliferating (CFSE(lo) ) CD4(+) T cells by flow cytometry. RESULTS: The frequency of tetramer(+) CD4(+) T cells determined ex vivo in PBMC of mugwort-allergic individuals ranged from 0 to 0.029%. After 2-3 weeks of in vitro expansion, sufficient tetramer(+) T cells for phenotyping were detected in 83% of Art v 125-36 -reactive T-cell lines (TCL) from mugwort-allergic individuals, but not in TCL from healthy individuals. The tetramers defined bona fide Th2 cells. Notably, Art v 125-36 -reactive TCL depleted of tetramer(+) T cells still reacted to the peptide, and only 44% of Art v 125-36 -specific T-cell clones were detected by the tetramer. CFSE(lo) CD4(+) T cells contained only 0.3-10.7% of tetramer(+) T cells and very low proportions of Th2 cells. CONCLUSION: Allergen-specific T cells can be identified by HLA class II tetramers with high specificity, but unexpected low sensitivity. In contrast, allergen-stimulated CFSE(lo) CD4(+) T cells contain extremely high fractions of bystander cells. Therefore, for T-cell monitoring, either method should be interpreted with caution.


Subject(s)
Allergens/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Peptides/immunology , Protein Multimerization/immunology , Amino Acid Sequence , Antigens, Plant/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/chemistry , Humans , Immunophenotyping , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Molecular Sequence Data , Peptides/chemistry , Phenotype , Pollen/adverse effects , Rhinitis, Allergic, Seasonal/diagnosis , Rhinitis, Allergic, Seasonal/immunology , T-Cell Antigen Receptor Specificity/immunology
17.
J Exp Med ; 210(10): 1961-76, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23999499

ABSTRACT

Dendritic cells (DCs) comprise distinct populations with specialized immune-regulatory functions. However, the environmental factors that determine the differentiation of these subsets remain poorly defined. Here, we report that retinoic acid (RA), a vitamin A derivative, controls the homeostasis of pre-DC (precursor of DC)-derived splenic CD11b(+)CD8α(-)Esam(high) DCs and the developmentally related CD11b(+)CD103(+) subset within the gut. Whereas mice deprived of RA signaling significantly lost both of these populations, neither pre-DC-derived CD11b(-)CD8α(+) and CD11b(-)CD103(+) nor monocyte-derived CD11b(+)CD8α(-)Esam(low) or CD11b(+)CD103(-) DC populations were deficient. In fate-tracking experiments, transfer of pre-DCs into RA-supplemented hosts resulted in near complete conversion of these cells into the CD11b(+)CD8α(-) subset, whereas transfer into vitamin A-deficient (VAD) hosts caused diversion to the CD11b(-)CD8α(+) lineage. As vitamin A is an essential nutrient, we evaluated retinoid levels in mice and humans after radiation-induced mucosal injury and found this conditioning led to an acute VAD state. Consequently, radiation led to a selective loss of both RA-dependent DC subsets and impaired class II-restricted auto and antitumor immunity that could be rescued by supplemental RA. These findings establish a critical role for RA in regulating the homeostasis of pre-DC-derived DC subsets and have implications for the management of patients with immune deficiencies resulting from malnutrition and irradiation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Homeostasis/immunology , Tretinoin/metabolism , Animals , Cell Differentiation/immunology , Cell Proliferation , Cell Survival , Dendritic Cells/cytology , Dendritic Cells/radiation effects , Female , Histocompatibility Antigens Class II/immunology , Humans , Immunophenotyping , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/radiation effects , Mice , Neoplasms/immunology , Neoplasms/metabolism , Organ Specificity/immunology , Phenotype , Receptors, Retinoic Acid/metabolism , Signal Transduction , Spleen/immunology , Spleen/metabolism , Spleen/radiation effects , Vitamin A/metabolism , Whole-Body Irradiation/adverse effects
18.
Am J Pathol ; 183(4): 1144-1155, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23911657

ABSTRACT

Immunization with human glucose-6-phosphate isomerase (hG6PI) protein or with several of its peptides induces arthritis in DBA/1 mice. We investigated G6PI peptide-induced arthritis in C57BL/10 mice and the effect of oxidative burst on disease. To study the arthritogenicity of G6PI peptides and its immune dependency, we used genetically modified and congenic mice on the C57BL/10 background and in vitro T- and B-cell assays. hG6PI(325-339) peptide induced arthritis in C57BL/10 mice. The disease was associated with major histocompatibility complex class II and was dependent on T cells, B cells, and complement C5. Th1 and Th17 cells primed with the hG6PI(325-339) peptide cross-reacted with the murine G6PI protein. The severity of the disease increased in mice carrying a mutation in Ncf1 (Ncf1*/*), which abolishes the NADPH oxidase 2 complex oxidative burst. Ncf1*/* mice developed arthritis also on immunization with the mouse G6PI325-339 peptide and in the absence of C5. The antibody responses to the G6PI protein and peptides were minimal in both Ncf1*/* and wild-type mice. Herein is described G6PI peptide as the first peptide to induce arthritis in C57BL/10 mice. The differences between the wild-type and Ncf1*/* mice suggest that an alternative complement-independent arthritogenic pathway could be operative in the absence of oxidative burst.


Subject(s)
Arthritis, Experimental/immunology , B-Lymphocytes/immunology , Complement System Proteins/immunology , Glucose-6-Phosphate Isomerase/immunology , Peptides/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , Amino Acids/genetics , Animals , Antigen Presentation/immunology , Arthritis, Experimental/blood , Arthritis, Experimental/chemically induced , Arthritis, Experimental/pathology , Chronic Disease , Cross-Priming/immunology , Cytokines/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , Mutation/genetics , NADPH Oxidases/metabolism , Protein Binding/immunology , Rats , Respiratory Burst/immunology , Th1 Cells/immunology , Th17 Cells/immunology
19.
Allergy ; 67(5): 593-600, 2012 May.
Article in English | MEDLINE | ID: mdl-22188598

ABSTRACT

BACKGROUND: Several studies in mice have recently shown that basophils can act as antigen-presenting cells (APC) inducing Th2-mediated immune responses against parasites or protease allergens. The aim of this study was to investigate whether human basophils function as APC for the major birch pollen allergen Bet v 1. METHODS: Fluorescently labeled Bet v 1 was used to assess surface binding and internalization of allergen by basophils and different types of APC from birch pollen-allergic and nonallergic individuals. Sorted basophils were analyzed in terms of up-regulation of MHC class II and co-stimulatory molecules in the absence and presence of IL-3 and IFN-γ by flow cytometry. Expression of proteins crucial for antigen presentation, namely cathepsin S and invariant chain, was determined. Basophils were used as APC in co-culture experiments with Bet v 1-specific T-cell clones (TCCs). RESULTS: Basophils from birch pollen-allergic donors very efficiently bound Bet v 1 through IgE/FcεRI complexes on their surface. In contrast to professional APC, basophils did not internalize allergen and expressed marginal levels of cathepsin S and invariant chain. HLA-DP, HLA-DQ, CD80/CD86, and CD40 were absent from purified basophils even when stimulated with IL-3 plus IFN-γ. IL-3/IFN-γ marginally up-regulated HLA-DR. Bet v 1-pulsed basophils failed to induce proliferative and cytokine responses in Bet v 1-specific, HLA-DR-restricted TCCs. CONCLUSION: Human basophils neither internalize, process nor present Bet v 1. Because Bet v 1 is a highly relevant allergen, we conclude that basophils play no role as APC in IgE-mediated allergy in humans.


Subject(s)
Allergens/immunology , Antigen-Presenting Cells/immunology , Antigens, Plant/immunology , Basophils/immunology , Pollen/immunology , Antigens, Plant/metabolism , Basophils/metabolism , Endocytosis/immunology , Epitopes/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Protein Binding/immunology , Receptors, IgE/immunology , Receptors, IgE/metabolism , T-Lymphocytes/immunology
20.
Clin J Am Soc Nephrol ; 6(11): 2573-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921153

ABSTRACT

BACKGROUND AND OBJECTIVES: In 2009, the pandemic influenza A/H1N1 accounted for worldwide recommendations about vaccination. There are few data concerning the immunogenicity or the security of the adjuvanted-A/H1N1 vaccine in transplanted and hemodialyzed patients. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Sera from 21 controls, 53 hemodialyzed (HD) patients, and 111 renal transplant recipients (RT) were sampled before (T0) and 1 month after (T1) a single dose of Pandemrix® vaccine (GSK Biologicals, AS03-adjuvanted). We measured the neutralizing antibodies against A/H1N1/2009, the geometric mean (GM) titers, the GM titer ratios (T1/T0) with 95% confidence intervals, and the seroconversion rate (responders: ≥4-fold increase in titer). The HLA and MICA immunization was determined by Luminex technology. RESULTS: The GM titer ratio was 38 (19 to 78), 9 (5 to 16), and 5 (3 to 6) for controls, HD patients, and RT patients, respectively (P < 0.001). The proportion of responders was 90%, 57%, and 44%, respectively (P < 0.001). In RT patients, the prevalence of histocompatibility leukocyte antigen (HLA) class I, histocompatibility leukocyte antigen class II, and MHC class I-related chain A immunization, was, respectively, 15%, 14%, and 14% before and 14%, 14%, and 11% after vaccination (P = 1, 1, and 0.39). CONCLUSIONS: The influenza A/H1N1-adjuvanted vaccine is of limited efficacy but is safe in renal disease populations. The humoral response is lower in transplanted versus hemodialyzed patients. Further studies are needed to improve the efficacy of vaccination in those populations.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Kidney Failure, Chronic/therapy , Kidney Transplantation , Renal Dialysis , Adult , Aged , Analysis of Variance , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Belgium , Case-Control Studies , Chi-Square Distribution , Cohort Studies , Drug Combinations , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunosuppressive Agents/administration & dosage , Influenza Vaccines/adverse effects , Influenza, Human/immunology , Influenza, Human/virology , Kidney Failure, Chronic/immunology , Kidney Failure, Chronic/surgery , Male , Middle Aged , Odds Ratio , Polysorbates/administration & dosage , Regression Analysis , Squalene/administration & dosage , Time Factors , Treatment Outcome , alpha-Tocopherol/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL