Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Monoclon Antib Immunodiagn Immunother ; 40(2): 52-59, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33900821

ABSTRACT

DNAM-1 is an activating immunoreceptor expressed on hematopoietic cells, including both CD4+ and CD8+ T cells, natural killer cells, and platelets. Since DNAM-1 is involved in the pathogenesis of various inflammatory diseases and cancers in humans as well as mouse models, it is a potential target for immunotherapy for these diseases. In this study, we generated a humanized neutralizing antihuman DNAM-1 monoclonal antibody (mAb), named TNAX101A, which contains an engineered Fc portion of human IgG1 to reduce Fc-mediated effector functions. We show that TNAX101A efficiently interfered the binding of DNAM-1 to its ligand CD155 and showed unique functions; it decreased production of the inflammatory cytokines such as interferon-gamma, tumor necrosis factor alpha, interleukin (IL)-6, IL-17A, and IL-17F by anti-CD3 antibody-stimulated or alloantigen-stimulated T cells and increased FOXP3 expression in anti-CD3-stimulated regulatory T (Treg) cells. These dual functions of TNAX101A may be advantageous for the treatment of T cell-mediated inflammatory diseases through both downregulation of effector T cell function and upregulation of Treg cell function.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Forkhead Transcription Factors/genetics , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized/genetics , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/genetics , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation/immunology , Humans , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunotherapy/trends , Neoplasms/immunology , Neoplasms/therapy , Th1 Cells/immunology , Th17 Cells/immunology
2.
Alcohol Clin Exp Res ; 45(5): 934-947, 2021 05.
Article in English | MEDLINE | ID: mdl-33704802

ABSTRACT

BACKGROUND: Chronic alcohol consumption is associated with a compromised innate and adaptive immune responses to infectious disease. Mucosa-associated invariant T (MAIT) cells play a critical role in antibacterial host defense. However, whether alcohol-associated deficits in innate and adaptive immune responses are mediated by alterations in MAIT cells remains unclear. METHODS: To investigate the impact of alcohol on MAIT cells, mice were treated with binge-on-chronic alcohol for 10 days and sacrificed at day 11. MAIT cells in the barrier organs (lung, liver, and intestine) were characterized by flow cytometry. Two additional sets of animals were used to examine the involvement of gut microbiota on alcohol-induced MAIT cell changes: (1) Cecal microbiota from alcohol-fed (AF) mice were adoptive transferred into antibiotic-pretreated mice and (2) AF mice were treated with antibiotics during the experiment. MAIT cells in the barrier organs were measured via flow cytometry. RESULTS: Binge-on-chronic alcohol feeding led to a significant reduction in the abundance of MAIT cells in the barrier tissues. However, CD69 expression on tissue-associated MAIT cells was increased in AF mice compared with pair-fed (PF) mice. The expression of Th1 cytokines and the corresponding transcriptional factor was tissue specific, showing downregulation in the intestine and increases in the lung and liver in AF animals. Transplantation of fecal microbiota from AF mice resulted in a MAIT cell profile aligned to that of AF mouse donor. Antibiotic treatment abolished the MAIT cell differences between AF and PF animals. CONCLUSION: MAIT cells in the intestine, liver, and lung are perturbed by alcohol use and these changes are partially attributable to alcohol-associated dysbiosis. MAIT cell dysfunction may contribute to alcohol-induced innate and adaptive immunity and consequently end-organ pathophysiology.


Subject(s)
Alcoholism/immunology , Binge Drinking/immunology , Central Nervous System Depressants/pharmacology , Dysbiosis/immunology , Ethanol/pharmacology , Gastrointestinal Microbiome , Mucosal-Associated Invariant T Cells/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Antigens, CD/drug effects , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Fecal Microbiota Transplantation , Flow Cytometry , Intestinal Mucosa/cytology , Lectins, C-Type/drug effects , Lectins, C-Type/metabolism , Liver/cytology , Liver/immunology , Lung/cytology , Lung/immunology , Mice , Mucosal-Associated Invariant T Cells/immunology
3.
Eur J Pharmacol ; 878: 173116, 2020 Jul 05.
Article in English | MEDLINE | ID: mdl-32315671

ABSTRACT

c-Jun N-terminal kinase (JNK) is a critical mitogen activated protein kinase (MAPK) implicated in inflammatory processes, with IQ-1S (11H-indeno[1,2-b]quinoxalin-11-one oxime sodium salt) being a high-affinity JNK inhibitor with pronounced anti-inflammatory properties. Here, we studied direct effects of IQ-1S on phenotypical and cytokine-producing characteristics of activated human monocytes/macrophages and T cells in vitro. Purified monocyte/macrophage cells were activated by bacterial lipopolysaccharide (LPS, 1 µg/ml) for 24 h, while T cells were activated by particles conjugated with antibodies (Abs) against human CD2, CD3, and CD28 for 48 h. Treatment with IQ-1S (0.5-25 µÐœ) in the presence of LPS reduced percentages of CD197 (CCR7)-positive cells in macrophage cultures, without affecting CD16+ (FcγRIII, low-affinity Fc-receptor), CD119+ (interferon-γ receptor 1), and CD124+ (IL-4 receptor α-subunit) cells. In addition, IQ-1S reduced production of tumour necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), IL-6, and IL-10 in macrophage cultures. In activated T cell cultures, IQ-1S decreased CD25+ cell numbers in both CD4-positive and CD4-negative T cell compartments. Central memory СD45RA-/СD197+ and effector memory СD45RA-/СD197- T cells were more sensitive to IQ-1S-mediated suppression, as compared to naïve СD45RA+/СD197+ and terminally-differentiated effector СD45RA+/СD197- T cells. IQ-1S also suppressed T-cell cytokine production (IL-2, interferon-É£, IL-4, and IL-10). Collectively, the results suggest that both human macrophage and T cells could be immediate cell targets for IQ-1S-based anti-inflammatory immunotherapy. IQ-1S-mediated suppressive effects were unlikely to be associated with macrophage/T helper polariation.


Subject(s)
Cell Differentiation/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Macrophages/drug effects , Oximes/pharmacology , Peptides/pharmacology , Phenylacetates/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinoxalines/pharmacology , T-Lymphocytes/drug effects , Adult , Antigens, Differentiation, T-Lymphocyte/drug effects , Blood/metabolism , Cytokines/metabolism , Drug Discovery , Female , Humans , Immunotherapy/methods , Lipopolysaccharides/metabolism , Lymphocyte Activation/drug effects , Male , Monocytes/drug effects , Phenotype , Receptors, Fc/metabolism , Receptors, Interferon/metabolism , Time Factors
4.
Clin Immunol ; 187: 76-84, 2018 02.
Article in English | MEDLINE | ID: mdl-29061445

ABSTRACT

NK cells represent a first line of immune defense, but are progressively dysregulated in multiple myeloma (MM) patients. To restore and facilitate their antitumor effect, NK cells are required in sufficient quantities and must be stimulated. We initially assessed the proportions of NKT and NK cells in 34 MM patients. The frequencies of both in PBMC populations correlated with those in BMMNCs irrespective of low BMMNC numbers. We then assessed the adjunctive effect of stimulating NKT cells with CD1d and α-GalCer complexes on the NK cells. The expression of NKG2D on CD56dimCD16+ NK cells and DNAM-1 on CD56brightCD16- NK cells increased after NKT cell activation. Apparently, NK cell-mediated anti-tumor effects were dependent on NKG2D and DNAM-1 ligands on myeloma cells. Thus, NK cell function in patients could be ameliorated, beyond the effect of immunosuppression, by NKT cell activation. This NKT-driven NK cell therapy could represent a potential new treatment modality.


Subject(s)
Killer Cells, Natural/immunology , Multiple Myeloma/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Adult , Aged , Aged, 80 and over , Antigens, CD1d/pharmacology , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Antineoplastic Agents/therapeutic use , Female , Galactosylceramides/pharmacology , Glucocorticoids/therapeutic use , Humans , Killer Cells, Natural/drug effects , Lymphocyte Activation , Male , Middle Aged , Multiple Myeloma/drug therapy , NK Cell Lectin-Like Receptor Subfamily K/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Natural Killer T-Cells/drug effects
5.
J Microbiol Biotechnol ; 27(6): 1189-1197, 2017 Jun 28.
Article in English | MEDLINE | ID: mdl-28372035

ABSTRACT

Lichen-forming fungi are known to have various biological activities, such as antioxidant, antimicrobial, antitumor, antiviral, anti-inflammation, and anti proliferative effects. However, the immunosuppressive effects of Bryoria sp. extract (BSE) have not previously been investigated. In this study, the inhibitory activity of BSE on the proliferation of CD8+ T cells and the mixed lymphocytes reaction (MLR) was evaluated in vitro. BSE was non-toxic in spleen cells and suppressed the growth of splenocytes induced by anti-CD3. The suppressed cell population in spleen cells consisted of CD8+ T cells and their proliferation was inhibited by the treatment with BSE. This extract significantly suppressed the IL-2 associated with T cell growth and IFN-γ as the CD8+ T cell marker. Furthermore, BSE reduced the expression of the IL-2 receptor alpha chain (IL-2Rα) on CD8+ T cells and CD86 on dendritic cells by acting as antigen-presenting cells. Finally, the MLR produced by the co-culture of C57BL/6 and MMC-treated BALB/c was suppressed by BSE. IL-2, IFN-γ, and CD69 on CD8+ T cells in MLR condition were inhibited by BSE. These results indicate that BSE inhibits the MLR via the suppression of IL-2Rα expression in CD8+ T cells. BSE has the potential to be developed as an anti-immunosuppression agent for organ transplants.


Subject(s)
Ascomycota/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Immunosuppressive Agents/pharmacology , Interleukin-2/biosynthesis , Animals , Antigens, CD/drug effects , Antigens, Differentiation, T-Lymphocyte/drug effects , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cell Proliferation/drug effects , Complex Mixtures/isolation & purification , Complex Mixtures/pharmacology , Cytotoxicity, Immunologic , Dendritic Cells/drug effects , Dendritic Cells/immunology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/immunology , Immunosuppressive Agents/isolation & purification , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-2/antagonists & inhibitors , Interleukin-2/immunology , Lectins, C-Type/drug effects , Lichens/chemistry , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Spleen/cytology , Spleen/drug effects , Spleen/immunology
6.
BMC Pharmacol Toxicol ; 16: 12, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25934555

ABSTRACT

BACKGROUND: Cytarabine is used in the treatment of acute myeloid leukemia (AML). Low-dose cytarabine can be combined with valproic acid and all-trans retinoic acid (ATRA) as AML-stabilizing treatment. We have investigated the possible risk of immunotoxicity by this combination. We examined the effects of cytarabine combined with valproic acid and ATRA on in vitro activated human T cells, and we tested cytarabine at concentrations reached during in vivo treatment with high doses, conventional doses and low doses. METHODS: T cells derived from blood donors were activated in vitro in cell culture medium alone or supplemented with ATRA (1 µM), valproic acid (500 or 1000 µM) or cytarabine (0.01-44 µM). Cell characteristics were assessed by flow cytometry. Supernatants were analyzed for cytokines by ELISA or Luminex. Effects on primary human AML cell viability and proliferation of low-dose cytarabine (0.01-0.5 µM) were also assessed. Statistical tests include ANOVA and Cluster analyses. RESULTS: Only cytarabine 44 µM had both antiproliferative and proapoptotic effects. Additionally, this concentration increased the CD4:CD8 T cell ratio, prolonged the expression of the CD69 activation marker, inhibited CD95L and heat shock protein (HSP) 90 release, and decreased the release of several cytokines. In contrast, the lowest concentrations (0.35 and 0.01 µM) did not have or showed minor antiproliferative or cytotoxic effects, did not alter activation marker expression (CD38, CD69) or the release of CD95L and HSP90, but inhibited the release of certain T cell cytokines. Even when these lower cytarabine concentrations were combined with ATRA and/or valproic acid there was still no or minor effects on T cell viability. However, these combinations had strong antiproliferative effects, the expression of both CD38 and CD69 was altered and there was a stronger inhibition of the release of FasL, HSP90 as well as several cytokines. Cytarabine (0.01-0.05 µM) showed a dose-dependent antiproliferative effect on AML cells, and in contrast to the T cells this effect reached statistical significance even at 0.01 µM. CONCLUSIONS: Even low levels of cytarabine, and especially when combined with ATRA and valproic acid, can decrease T cell viability, alter activation-induced membrane-molecule expression and decrease the cytokine release.


Subject(s)
Cytarabine/pharmacology , Drug Interactions , Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , Tretinoin/pharmacology , Valproic Acid/pharmacology , ADP-ribosyl Cyclase 1/biosynthesis , ADP-ribosyl Cyclase 1/drug effects , Antigens, CD/biosynthesis , Antigens, CD/drug effects , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/drug effects , CD4 Antigens/immunology , CD8 Antigens/immunology , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Proliferation , Cell Survival/drug effects , Cells, Cultured , Cytarabine/administration & dosage , Cytokines/metabolism , Dose-Response Relationship, Drug , Fas Ligand Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Lectins, C-Type/biosynthesis , Lectins, C-Type/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tretinoin/administration & dosage
7.
Immunopharmacol Immunotoxicol ; 37(1): 42-55, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25347443

ABSTRACT

CONTEXT: Saponinum album (SA) is a complex mixture of triterpenoid saponins previously shown to augment the cytotoxicity of the type I ribosome-inactivating protein saporin and an EGF-saporin target toxin that could potentially be used to improve the therapeutic window of targeted toxins. OBJECTIVE: To investigate the augmentative property of SA on saporin and saporin-based immunotoxins (IT) directed against five different cell surface target molecules on human leukemia and lymphoma cells. MATERIALS AND METHODS: After determining the optimum dose of SA for each cell line, the extent of SA-mediated augmentation was established for saporin and five saporin-based ITs using XTT and an annexin V apoptosis assay. Immunospecificity was investigated using three different blocking assays. Dose-scheduling was also investigated using the XTT assay. RESULTS: Uncorrected SA-mediated augmentation ranged at best from 31.5 million-fold to, at worse, 174-fold. However, when the calculated fold-increases were adjusted for the non-immunospecific effects of SA on an off-target IT, the true augmentative effects of SA were found to be largely non-immunospecific. Antibody blocking studies demonstrated that the augmentative effect of SA was only partially immunospecific. Separate exposure of target cells to IT and SA at different times demonstrated that immunospecific augmentation of IT by SA could be achieved but only if cells were exposed to IT first and SA second. CONCLUSIONS: SA significantly, although variably, augments the cytotoxicity of saporin and saporin-based immunotoxins. Concomitant exposure to both IT and SA can result in non-immunospecific cytotoxicity that can be overcome by temporally separating exposure to each.


Subject(s)
Apoptosis/drug effects , Cytotoxicity, Immunologic/drug effects , Ribosome Inactivating Proteins, Type 1/pharmacology , Saponins/pharmacology , Triterpenes/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/immunology , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/immunology , Burkitt Lymphoma/immunology , Burkitt Lymphoma/pathology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Administration Schedule , Flow Cytometry , Humans , Molecular Targeted Therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Ribosome Inactivating Proteins, Type 1/administration & dosage , Ribosome Inactivating Proteins, Type 1/isolation & purification , Saponins/administration & dosage , Saponins/isolation & purification , Saporins , Triterpenes/administration & dosage , Triterpenes/isolation & purification
8.
Iran J Allergy Asthma Immunol ; 14(5): 519-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26742441

ABSTRACT

Tetanus is caused by the tetanus neurotoxin (TeNT), a 150 kDa single polypeptide molecule which is cleaved into active two-chain molecules composed of a 50 kDa N-terminal light (L) and a 100 kDa C-terminal heavy (H) chains. Fragment C is further subdivided into two subdomains: the proximal HCN  subdomain and the extreme carboxy subdomain, HCC. HCC is considered as an immunodominant part of TeNT and is responsible for TeNT binding activity to neurons.In the present study, we investigated the ability of recombinant HCC(r HCC) to induce T cell activation. Our results showed that recombinant HCC has a stimulatory effect on IFN-γ secretion by T cells after 48h co-incubation in the presence of anti-TLR-2 Ab. Also, Hcc can induce the expression of CD69 on T cells.Our finding indicated that stimulatory effects of HCC on T cells are TLR-2 independent and anti-TLR-2 inhibitory antibody fails to neutralize HCC stimulatory effects on T cells.Furthermore, HCC  is critical for immunogenic activity of TeNT and is able to induce T cells through TLR-2 independent pathway.


Subject(s)
Interferon-gamma/drug effects , Lymphocyte Activation/drug effects , Neuromuscular Blocking Agents/pharmacology , Peptide Fragments/pharmacology , T-Lymphocytes/drug effects , Tetanus Toxin/pharmacology , Tumor Necrosis Factor-alpha/drug effects , Antibodies/immunology , Antibodies/pharmacology , Antigens, CD/drug effects , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/immunology , Humans , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lectins, C-Type/drug effects , Lectins, C-Type/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptor 2/immunology , Tumor Necrosis Factor-alpha/metabolism
9.
J Dent Res ; 93(11): 1095-100, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25252878

ABSTRACT

Semaphorin 4d (Sema4d) has been proposed as a novel target gene for the treatment of osteoporosis. Recently, we fabricated a site-specific bone-targeting system from polymeric nanoparticles that demonstrates an ability to prevent bone loss in an osteoporotic model by interfering with Sema4d gene expression using small interference RNA (siRNA) molecules. The aim of the present investigation was to determine the effects of this targeting system on the periodontium, an area of high bone turnover. We demonstrated, by single photon emission computed tomography, that intravenous injection of this molecule in ovariectomized Balb/C mice is able to target alveolar bone peaking 4 hr post-injection. We then compared, by histological analysis, the bone volume/total volume (BV/TV), alveolar bone height loss, immunohistochemical expression of Sema4d, and total number of osteoclasts in mandibular alveolar bone. Four treatment modalities were compared as follows: (1) sham-operated, (2) OVX-operated, (3) OVX+estrogen replacement therapy, and (4) OVX+siRNA-Sema4d animals. The results from the present study demonstrate that an osteoporotic condition significantly increases alveolar bone height loss, and that the therapeutic effects via bone-targeting systems featuring interference of Sema4d are able to partly counteract alveolar bone loss caused by osteoporosis. While the future therapeutic demand for the large number of patients suffering from osteoporosis faces many challenges, we demonstrate within the present study an effective drug-delivery moiety with anabolic effects on the bone remodeling cycle able to locate and target alveolar bone regeneration.


Subject(s)
Alveolar Bone Loss/prevention & control , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/genetics , Gene Silencing , Lymphocyte Activation/genetics , Osteoporosis/prevention & control , Semaphorins/genetics , Alveolar Process/pathology , Animals , Antigens, Differentiation, T-Lymphocyte/drug effects , Bone Regeneration/genetics , Bone Remodeling/genetics , Disease Models, Animal , Estrogen Replacement Therapy/methods , Female , Injections, Intravenous , Lymphocyte Activation/drug effects , Mandible/pathology , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Organ Size , Osteoclasts/pathology , Ovariectomy/methods , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Semaphorins/antagonists & inhibitors , Tomography, Emission-Computed, Single-Photon/methods , X-Ray Microtomography/methods
10.
Scand J Med Sci Sports ; 22(2): 249-58, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22404431

ABSTRACT

This study investigated the effect of caffeine ingestion on antigen-stimulated T- (CD4(+) and CD8(+) ) and natural killer (NK)- (CD3(-) CD56(+) ) cell activation after prolonged, strenuous cycling. In a randomized cross-over design, nine male endurance cyclists (age: 22 ± 3 years, VO(2peak) : 62 ± 4 mL/kg/min, mean ± SD) cycled for 90 min at 70% VO(2peak) 60 min after ingesting 6 mg/kg body mass of caffeine (CAF) or placebo (PLA). Venous blood samples were obtained before supplementation, pre-exercise, immediately post-exercise and 1 h post-exercise. Whole blood was stimulated with Pediacel (five in one) vaccine. At 1 h post-exercise the number of antigen-stimulated CD4(+) cells expressing CD69 decreased on CAF compared with PLA [15 (17) × 10(6) vs 23 (22) × 10(6) cells/L, P<0.05]. In addition, the geometric mean fluorescence intensity (GMFI) of CD69 expression on antigen-stimulated CD8(+) cells decreased on CAF compared with PLA 1 h post-exercise [78 (10)% vs 102 (24)%, P<0.05]. At the same time-point GMFI of CD69 expression on antigen-stimulated CD3(-) CD56(+) cells was increased on CAF compared with PLA [103 (9)% vs 87 (8)%, P<0.05]. These findings suggest that caffeine reduces antigen-stimulated CD69 expression on T cells while at the same time increases NK-cell activation 1 h after intensive cycling.


Subject(s)
Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , T-Lymphocytes/drug effects , Adult , Antigens, CD/drug effects , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Bicycling/physiology , Caffeine/immunology , Central Nervous System Stimulants/immunology , Cross-Over Studies , Exercise/physiology , Humans , Killer Cells, Natural/immunology , Lectins, C-Type/drug effects , Lectins, C-Type/metabolism , Male
11.
Exp Dermatol ; 21(3): 228-30, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22379972

ABSTRACT

8-Methoxypsoralen plus UVA (PUVA) photochemotherapy is an effective treatment for many skin diseases including psoriasis. However, its exact mechanism of therapeutic action is incompletely understood. Previously, in K5.hTGFß1 transgenic psoriatic mice, we found that PUVA induces Foxp3+ CD25+ CD4+ regulatory T cells in both lymph node and spleen. Now, in the same model, we investigated whether cutaneous lymphocyte-associated antigen (CLA) mediates PUVA's effect on homing of CD25+ CD4+ T cells to the lymph nodes of K5.hTGFß1 transgenic mice. We found that a low dose of topical PUVA maximally increased the proportion of CLA + CD25+ CD4 + T cells in the lymph nodes by up to 8-fold. We also observed an increased number of Foxp3+ CD25+ T cells in the skin of the mice after PUVA treatment. Together, these findings suggest that PUVA affects the homing of regulatory T cells.


Subject(s)
Lymph Nodes/cytology , Methoxsalen/administration & dosage , PUVA Therapy , Photosensitizing Agents/administration & dosage , Psoriasis/drug therapy , T-Lymphocytes/drug effects , Animals , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/radiation effects , Disease Models, Animal , Interleukin-2 Receptor alpha Subunit/drug effects , Interleukin-2 Receptor alpha Subunit/radiation effects , Lymph Nodes/drug effects , Lymph Nodes/radiation effects , Mice , Mice, Transgenic , Psoriasis/immunology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/radiation effects
12.
Semin Oncol ; 37(5): 473-84, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21074063

ABSTRACT

Cytotoxic T-lymphocyte-associated antigen (CTLA-4), also known as CD152, is a co-inhibitory molecule that functions to regulate T-cell activation. Antibodies that block the interaction of CTLA-4 with its ligands B7.1 and B7.2 can enhance immune responses, including antitumor immunity. Two CTLA-4-blocking antibodies are presently under clinical investigation: ipilimumab and tremelimumab. CTLA-4 blockade has shown promise in treatment of patients with metastatic melanoma, with a recently completed randomized, double-blind phase III trial demonstrating a benefit in overall survival (OS) in the treated population. However, this approach appears to benefit only a subset of patients. Understanding the mechanism(s) of action of CTLA-4 blockade and identifying prognostic immunologic correlates of clinical endpoints to monitor are presently areas of intense investigation. Several immunologic endpoints have been proposed to correlate with clinical activity. This review will focus on the endpoints of immune monitoring described in studies to date and discuss future areas of additional work needed.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antineoplastic Agents/immunology , Melanoma/drug therapy , Monitoring, Immunologic , Skin Neoplasms/drug therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antigens, Differentiation, T-Lymphocyte/blood , Antigens, Differentiation, T-Lymphocyte/drug effects , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Biomarkers , CTLA-4 Antigen , HLA-DR Antigens/blood , HLA-DR Antigens/drug effects , Humans , Inducible T-Cell Co-Stimulator Protein , Ipilimumab , Leukocyte Common Antigens/blood , Leukocyte Common Antigens/drug effects , Lymphocyte Count , Melanoma/immunology , Monitoring, Immunologic/methods , Skin Neoplasms/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
13.
Cancer Res ; 70(20): 7960-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20798218

ABSTRACT

Inflammatory bowel diseases (IBD) increase the risk of developing colorectal cancer. Dietary components that reduce inflammation are associated with lower cancer risk. The long-chain omega-3 fatty acid docosahexaenoic acid (DHA) is present in fish oil and has potent anti-inflammatory properties. The objective of this study is to determine whether dietary fish oil enriched with DHA (DFO) could reduce experimentally induced colitis and colon cancer risk in a mouse model. When SMAD3-/- mice are exposed to Helicobacter hepaticus, mild colitis is observed 4 weeks postinfection. Mice were fed isocaloric diets modified to include corn oil, safflower oil, or DFO (doses ranging from 0.75% to 6.00%) as the fatty acid source for 8 weeks. Mice were gavaged with H. hepaticus; DFO feeding was continued; and mice were sacrificed 4 weeks after infection. The colon and cecum were collected for histopathology. Spleens and mesenteric lymph nodes were collected and analyzed for T-cell populations using flow cytometry. Contrary to expectations, DFO induced severe colitis and adenocarcinoma formation. DFO consumption was associated with decreased CD8(+) cell frequency and diminished CD69 expression on CD4(+) and CD8(+) T-cell populations. Mice consuming DFO also exhibited higher FoxP3(+) CD25(+) CD4(+) T regulatory cell frequency, FoxP3 expression, and altered L-selectin expression during infection. We concluded that DFO-fed mice may be less equipped to mount a successful response to H. hepaticus infection, increasing colon cancer risk. These results support the need to establish a tolerable upper limit for DHA intake particularly in the context of chronic inflammatory conditions such as IBD.


Subject(s)
Colitis/immunology , Dietary Fats, Unsaturated/pharmacology , Fish Oils/pharmacology , Inflammation/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD/drug effects , Antigens, Differentiation, T-Lymphocyte/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Colitis/chemically induced , Colitis/pathology , Colitis/prevention & control , Colon/drug effects , Colon/immunology , Colonic Neoplasms/prevention & control , Corn Oil/pharmacology , Disease Models, Animal , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Fish Oils/administration & dosage , Flow Cytometry , Inflammation/pathology , Lectins, C-Type/drug effects , Mice , Mice, Knockout , Rectum/drug effects , Rectum/immunology , Smad3 Protein/deficiency , Smad3 Protein/genetics , T-Lymphocytes/drug effects
14.
J Neuroimmunol ; 220(1-2): 52-63, 2010 Mar 30.
Article in English | MEDLINE | ID: mdl-20149931

ABSTRACT

Rapamycin is an oral immunosuppressant drug previously reported to efficiently induce naturally occurring CD4(+)CD25(+)FoxP3(+) regulatory T ((n)T(reg)) cells re-establishing long-term immune self-tolerance in autoimmune diseases. We investigated the effect of rapamycin administration to SJL/j mice affected by PLP(139-151)-induced relapsing-remitting experimental autoimmune encephalomyelitis (RR-EAE). We found that oral or intraperitoneal treatment at the peak of disease or at the end of the first clinical attack, dramatically ameliorated the clinical course of RR-EAE. Treatment suspension resulted in early reappearance of disease. Clinical response was associated with reduced central nervous system demyelination and axonal loss. Rapamycin induced suppression of IFN-gamma, and IL-17 release from antigen-specific T cells in peripheral lymphoid organs. While CD4(+)FoxP3(+) cells were unaffected, we observed disappearance of CD4(+)CD45RB(high) effector T (T(eff)) cells and selective expansion of T(reg) cells bearing the CD4(+)CD45RB(low)FoxP3(+)CD25(+)CD103(+) extended phenotype. Finally, the dual action of rapamycin on both T(eff) and T(reg) cells resulted in modulation of their ratio that closely paralleled disease course. Our data show that rapamycin inhibits RR-EAE, provide evidence for the immunological mechanisms, and indicate this compound as a potential candidate for the treatment of multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Immune Tolerance/drug effects , Immunosuppressive Agents/pharmacology , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/drug effects , Animals , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Surface/immunology , Disease Models, Animal , Drug Administration Routes , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunosuppressive Agents/therapeutic use , Interferon-gamma/drug effects , Interferon-gamma/metabolism , Interleukin-17/metabolism , Mice , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/immunology , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Myelin Proteolipid Protein/pharmacology , Peptide Fragments/pharmacology , Phenotype , Secondary Prevention , Sirolimus/therapeutic use , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
15.
Br J Dermatol ; 162(6): 1206-15, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20163421

ABSTRACT

BACKGROUND: 1,24-Dihydroxyvitamin D3 (tacalcitol), a vitamin D(3) compound, has been used to treat T cell-mediated inflammatory skin diseases such as psoriasis, prurigo and vitiligo. The best-known mechanism of action of this compound is inhibition of the abnormal proliferation of keratinocytes and subsequent maturation; however, its effects on skin T-cell recruitment have not yet been evaluated. Cutaneous lymphocyte-associated antigen (CLA), a surface glycoprotein expressed on T cells, plays a critical role in skin T-cell infiltration. We recently reported that 1,25-dihydroxyvitamin D3 inhibits skin infiltration of CD4+ T cells by suppressing CLA expression on T cells. OBJECTIVES: In this study, we investigated the effect of tacalcitol on CLA epitope decoration and on the levels of gut or lymph node homing receptor expression in human T cells. METHODS: We cultured human T cells with tacalcitol and analysed the effect on CLA expression and skin-homing ability, and evaluated glycosyltransferase mRNAs. We also performed an in vivo study using an antigen-dependent delayed-type hypersensitivity (DTH) mouse model and investigated the effect of tacalcitol on skin-infiltrating CD4+ T cells. RESULTS: Tacalcitol downregulated the expression of CLA and, in parallel, the E- and P-selectin ligand function; however, it exerted no effect on other homing receptors. Subcutaneously and intraperitoneally administered tacalcitol downregulated skin infiltration of effector CD4+ T cells in an in vivo DTH mouse model. CONCLUSIONS: These findings suggest that tacalcitol reduces skin inflammation by partially downregulating CLA expression levels.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/drug effects , Cell Movement/drug effects , Dermatologic Agents/pharmacology , Dihydroxycholecalciferols/pharmacology , Membrane Glycoproteins/drug effects , Skin/immunology , T-Lymphocytes/drug effects , Adult , Animals , Antigens, Differentiation, T-Lymphocyte/metabolism , Dermatitis, Contact/immunology , Dermatitis, Contact/pathology , Disease Models, Animal , Down-Regulation , E-Selectin/metabolism , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Membrane Glycoproteins/metabolism , Mice , P-Selectin/metabolism , Receptors, Lymphocyte Homing/drug effects , Receptors, Lymphocyte Homing/metabolism , T-Lymphocytes/metabolism
16.
AIDS Res Hum Retroviruses ; 25(12): 1313-28, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20001520

ABSTRACT

Reports indicate that myeloid and plasmacytoid dendritic cells (mDCs and pDCs), which are key effector cells in host innate immune responses, can be infected with HIV-1 and are reduced in number and function during the chronic phase of HIV disease. Furthermore, it was recently demonstrated that a sustained loss of mDCs and pDCs occurs in SIV-infected macaques. Since loss of functional DC populations might impair innate immune responses to opportunistic microorganisms and neoplastic cells, we explored whether inoculation of naive and SIV- or SHIV-infected pigtailed macaques with the hematopoietic cytokine FLT3-ligand (FLT3-L) would expand the number of mDCs and pDCs in vivo. After the macaques received supraphysiologic doses of FLT3-L, mDCs, pDCs, and monocytes increased up to 45-fold in blood, lymph nodes, and bone marrow (BM), with DC expansion in the BM preceding mobilization in blood and lymphoid tissues. FLT3-L also increased serum levels of IL-12, at least transiently, and elicited higher surface expression of HLA-DR and the activation markers CD25 and CD69 on NK and T cells. During and after treatment of infected animals, APCs increased in number and were activated; however, CD4(+) T cell numbers, virion RNA, and anti-SIV/SHIV antibody titers remained relatively stable, suggesting that FLT3-L might be a safe modality to expand DC populations and provide therapeutic benefit during chronic lentivirus infections.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Dendritic Cells/drug effects , Killer Cells, Natural/drug effects , Membrane Proteins/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus , Adjuvants, Immunologic/pharmacology , Animals , Antigens, CD/drug effects , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/metabolism , B7-1 Antigen/drug effects , B7-1 Antigen/metabolism , B7-2 Antigen/drug effects , B7-2 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/agonists , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Female , Interferon-alpha/blood , Interleukin-12/agonists , Interleukin-12/blood , Interleukin-2 Receptor alpha Subunit/drug effects , Interleukin-2 Receptor alpha Subunit/metabolism , Killer Cells, Natural/immunology , Lectins, C-Type/drug effects , Lectins, C-Type/metabolism , Macaca nemestrina , Male , Tumor Necrosis Factor-alpha/blood
17.
Klin Padiatr ; 221(6): 344-50, 2009.
Article in English | MEDLINE | ID: mdl-19890785

ABSTRACT

Inducible co-stimulator (ICOS) interaction with its ligand (ICOSL) is involved in several T cell effector functions. While blockade of ICOS:ICOSL interaction in chronic graft versus host disease (GVHD) seems beneficial, results for acute GVHD remain controversial. To further elucidate its role in acute GVHD, C57BL/6 mice were reconstituted with allogeneic spleen cells in the absence or presence of ICOSL-blocking mAb. Mice reconstituted with allogeneic spleen cells experienced severe GVHD and died untreated within 6-9 days after transplantation. Mice treated with an anti-ICOSL mAb starting from day 3 after transplantation gained weight again and survived for at least additional 12 days, although the treatment was already stopped at day 11 after transplantation. In contrast, the anti-ICOSL treatment starting from day 0 did not prevent GVHD. The difference between therapeutic (day 3) and prophylactic (day 0) anti-ICOSL treatment was independent of CD25+CD4+ regulatory T cells since their depletion did not abrogate the therapeutic effect of ICOSL blockade. Microarray analysis revealed IFN-gamma and chemokine up-regulation in spleen cells of prophylactically treated mice, emphasizing kinetic dependence of acute GVHD modulation via blockade of ICOS:ICOSL interaction.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation, T-Lymphocyte/drug effects , Graft vs Host Disease/immunology , Proteins/antagonists & inhibitors , Animals , Bone Marrow Transplantation , Chemokines/metabolism , Drug Administration Schedule , Female , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Interferon-gamma/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Spleen/immunology , Spleen/transplantation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Up-Regulation
18.
Mol Immunol ; 46(15): 3108-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19592095

ABSTRACT

Both NK cells and IL-15 play crucial roles in innate immunity against viral infections and cancer. Cigarette smoke is known to increase susceptibility to infections and certain cancers. Interleukin (IL)-15 plays an important role in immune responses by regulating proliferation, survival and functions of NK cells. Here, we examined the impact of cigarette smoke on IL-15 production and IL-15 mediated NK cell functions in human PBMCs. We report that cigarette smoke significantly suppresses the induction of IL-15 by poly I:C in human PBMCs. Serum IL-15 levels among smokers was significantly lower than non-smokers. In contrast to a profound increases in intracellular IL-15/IL-15Ralpha in poly I:C-treated PBMCs, exposure of PBMCs to smoke-conditioned media (SCM) diminished the IL-15/IL-15Ralpha production. We examined if inhibition of IL-15 production could lead to less NK cell activation. Interestingly, SCM-treated PBMCs had diminished up-regulation of NK cell activation marker, CD69, but not NKG2D compared with controls after poly I:C stimulation. We then confirmed by using IL-15 neutralizing antibody as well as exogenous IL-15 that the ploy I:C-induced NK cells activation was IL-15 mediated. More importantly, cigarette smoke significantly impaired NK cell cytolytic potential to kill K562 cancer cells which was found to be IL-15 mediated. The inhibition of IL-15 and its regulatory NK cell activities were linked to attenuated STAT3 and STAT5, but not ERK1/2 phosphorylations. We demonstrate, for the first time, that cigarette smoke compromises IL-15 production and as a result NK cell function which could link to the higher incidence of cancers or viral infections observed among smokers.


Subject(s)
Interleukin-15/antagonists & inhibitors , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/immunology , Nicotiana/adverse effects , Smoke/adverse effects , Smoking/adverse effects , Antigens, CD/drug effects , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Culture Media, Conditioned/pharmacology , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interferon Inducers/pharmacology , Interleukin-15/blood , Interleukin-15 Receptor alpha Subunit/antagonists & inhibitors , Interleukin-15 Receptor alpha Subunit/immunology , Killer Cells, Natural/drug effects , Lectins, C-Type , Leukocytes, Mononuclear/drug effects , NK Cell Lectin-Like Receptor Subfamily K/drug effects , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Phosphorylation/drug effects , Phosphorylation/immunology , Poly I-C/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Smoking/blood , Smoking/immunology
19.
MAbs ; 1(5): 453-61, 2009.
Article in English | MEDLINE | ID: mdl-20065650

ABSTRACT

A costimulatory signal is required for the full activation of T cells, in addition to the antigen-specific signal via the T cell receptor. The inducible costimulator, ICOS is one of the costimulatory molecules that play an essential role in this process, particularly in the expansion or the development of effector T cells. As blocking of the interaction between ICOS and its ligand, B7RP-1, suppresses the T cell response, it can be applied to the treatment of allograft rejection or autoimmune diseases. Here, we isolated four scFv clones that were specific to human B7RP-1 by biopanning a human antibody phage library. We found that three of these clones inhibited the interaction between ICOS-Fc and B7RP-1-Fc. These inhibitory clones not only recognized B7RP-1 molecules expressed on B cells, as assessed by FACS, but also exhibited inhibitory activity in a proliferation assay of T cells stimulated with anti-CD3 mAb and B7RP-1-Fc. Finally, the suppression effect of the scFv on the allogenic immune response was examined using a mixed lymphocyte reaction assay, which demonstrated a successful inhibition of the allogenic reaction, in spite of the high dose needed for complete inhibition (360 nM).


Subject(s)
Antigens, Differentiation, T-Lymphocyte/drug effects , B7-1 Antigen/immunology , Immunoglobulin Fragments/immunology , Lymphocyte Activation , T-Lymphocytes , Amino Acid Sequence , Animals , Antibody Specificity , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Flow Cytometry , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Fragments/therapeutic use , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Molecular Sequence Data , Peptide Library , Surface Plasmon Resonance , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Transplantation, Homologous/immunology
20.
Cardiovasc Res ; 81(1): 197-205, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18703531

ABSTRACT

AIMS: Atherosclerosis is a chronic inflammatory disease regulated by immune mechanisms. CD69 is a cell surface receptor rapidly induced after leukocyte activation at sites of chronic inflammation. Genetic disruption of CD69 in the mouse aggravates collagen-induced arthritis (CIA), and partial depletion of CD69-expressing cells with anti-CD69 monoclonal antibody (mAb) prevents CIA development in wild-type mice, suggesting that this receptor negatively modulates immune and inflammatory responses. It has been recently reported that CD69 is upregulated in a large subset of T cells in atherosclerosis-prone apolipoprotein E-null mice (apoE(-/-)). In this study, we investigated whether altering CD69 function affects atherosclerosis development. METHODS AND RESULTS: We studied native and diet-induced atherosclerosis in apoE(-/-) and doubly deficient apoE(-/-)CD69(-/-) mice and performed expression studies in tissues and primary cells derived from these animals. Plasma cholesterol level was unaffected by CD69 genetic inactivation. Although this genetic manipulation led to an elevated production of interferon gamma and interleukin 10 by activated T cells, apoE(-/-) and apoE(-/-)CD69(-/-) mice fed control and high-fat diet exhibited atheromas of similar size and composition when analysed at different stages of the disease. Likewise, anti-CD69 mAb treatment had no effect on plasma cholesterol and atherosclerosis burden in fat-fed apoE(-/-) mice. CONCLUSION: In contrast to previous studies highlighting the protective function of CD69 against CIA, an autoimmune inflammatory disease, our results rule out a significant role for CD69 against atherosclerosis in apoE(-/-) mice, an experimental disease model featuring a local inflammatory response triggered and sustained by alterations in lipid homeostasis.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/drug effects , Antigens, CD/genetics , Antigens, Differentiation, T-Lymphocyte/drug effects , Antigens, Differentiation, T-Lymphocyte/genetics , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/physiopathology , Cholesterol/blood , Disease Models, Animal , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lectins, C-Type , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...