Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 62(7): e202210209, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36316282

ABSTRACT

Amyloid-ß (Aß) in the form of neurotoxic aggregates is regarded as the main pathological initiator and key therapeutic target of Alzheimer's disease. However, anti-Aß drug development has been impeded by the lack of a target needed for structure-based drug design and low permeability of the blood-brain barrier (BBB). An attractive therapeutic strategy is the development of amyloid-based anti-Aß peptidomimetics that exploit the self-assembling nature of Aß and penetrate the BBB. Herein, we designed a dimeric peptide drug candidate based on the N-terminal fragment of Aß, DAB, found to cross the BBB and solubilize Aß oligomers and fibrils. Administration of DAB reduced amyloid burden in 5XFAD mice, and downregulated neuroinflammation and prevented memory impairment in the Y-maze test. Peptide mapping assays and molecular docking studies were utilized to elucidate DAB-Aß interaction. To further understand the active regions of DAB, we assessed the dissociative activity of DAB with sequence modifications.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Molecular Docking Simulation , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/chemistry , Amyloid , Cognitive Dysfunction/drug therapy , Mice, Transgenic
2.
BMB Rep ; 51(6): 314, 2018 06.
Article in English | MEDLINE | ID: mdl-29941089

ABSTRACT

The BMB Reports would like to correct in the ACKNOWLEDGEMENTS of BMB Rep. 50(11): 578-583 titled "PD-1 deficiency protects experimental colitis via alteration of gut microbiota."

3.
BMB Rep ; 50(11): 578-583, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29065967

ABSTRACT

Programmed cell death-1 (PD-1) is a coinhibitory molecule and plays a pivotal role in immune regulation. Here, we demonstrate a role for PD-1 in pathogenesis of inflammatory bowel disease (IBD). Wild-type (WT) mice had severe wasting disease during experimentally induced colitis, while mice deficient for PD-1 (PD-1-/-) did not develop colon inflammation. Interestingly, PD-1-/- mice cohoused with WT mice became susceptible to colitis, suggesting that resistance of PD-1-/- mice to colitis is dependent on their gut microbiota. 16S rRNA gene-pyrosequencing analysis showed that PD-1-/- mice had altered composition of gut microbiota with significant reduction in Rikenellaceae family. These altered colon bacteria of PD-1-/- mice induced less amount of inflammatory mediators from colon epithelial cells, including interleukin (IL)-6, and inflammatory chemokines. Taken together, our study indicates that PD-1 expression is involved in the resistance to experimental colitis through altered bacterial communities of colon. [BMB Reports 2017; 50(11): 578-583].


Subject(s)
Colitis/metabolism , Colitis/prevention & control , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Animals , Bacteria/genetics , Colitis/chemically induced , Colitis/pathology , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Inflammatory Bowel Diseases/metabolism , Metagenomics/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Programmed Cell Death 1 Ligand 2 Protein/genetics , RNA, Ribosomal, 16S/genetics
4.
Immunol Lett ; 176: 36-43, 2016 08.
Article in English | MEDLINE | ID: mdl-27229912

ABSTRACT

IL-12p40 homodimer, a natural antagonist of IL-12 and IL-23, performs an important role in the expression of proinflammatory cytokines that is essential for Th1 and Th17 immune responses. Here, we reveal the therapeutic and immunosuppressive effect of the IL-12p40 subunit ((p40)2-Fc) in an experimental autoimmune arthritis model. We hypothesized that (p40)2-Fc may reduce the inflammatory response and the activation of T cells. In this study, we intraperitoneally injected (p40)2-Fc into collagen induced arthritis (CIA) mice to identify whether (p40)2-Fc attenuates CIA severity. (p40)2-Fc reduced the development of CIA, joint inflammation and cartilage destruction. (p40)2-Fc also significantly decreased the concentration of serum immunoglobulin as well as the number of T cells and C II specific T cells. In addition, osteoclastogenesis in (p40)2-Fc treated mice was down-regulated compared to the mice treated with (p40)2-Fc control. We observed that (p40)2-Fc treatment alleviates arthritis in mice with CIA, reducing inflammation and osteoclast differentiation. These findings suggest that (p40)2-Fc can be a potential therapeutic approach for autoimmune arthritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/therapy , Immunoglobulin Fc Fragments/therapeutic use , Immunotherapy/methods , Interleukin-12 Subunit p40/immunology , T-Lymphocytes/immunology , Animals , Antibody Formation/drug effects , Arthritis, Experimental/immunology , Cells, Cultured , Collagen Type II/immunology , Immunosuppression Therapy , Injections, Intraperitoneal , Male , Mice , Mice, Inbred DBA , Osteogenesis/drug effects
5.
J Virol ; 90(5): 2273-84, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26656713

ABSTRACT

UNLABELLED: Influenza A virus (IAV) infection frequently causes hospitalization and mortality due to severe immunopathology. Annual vaccination and antiviral drugs are the current countermeasures against IAV infection, but they have a limited efficacy against new IAV variants. Here, we show that intranasal pretreatment with Fc-fused interleukin-7 (IL-7-mFc) protects mice from lethal IAV infections. The protective activity of IL-7-mFc relies on transcytosis via neonatal Fc receptor (FcRn) in the lung and lasts for several weeks. Introduction of IL-7-mFc alters pulmonary immune environments, leading to recruitment of T cells from circulation and their subsequent residency as tissue-resident memory-like T (TRM-like) cells. IL-7-mFc-primed pulmonary TRM-like cells contribute to protection upon IAV infection by dual modes. First, TRM-like cells, although not antigen specific but polyclonal, attenuate viral replication at the early phase of IAV infection. Second, TRM-like cells augment expansion of IAV-specific cytotoxic T lymphocytes (CTLs), in particular at the late phase of infection, which directly control viruses. Thus, accelerated viral clearance facilitated by pulmonary T cells, which are either antigen specific or not, alleviates immunopathology in the lung and mortality from IAV infection. Depleting a subset of pulmonary T cells indicates that both CD4 and CD8 T cells contribute to protection from IAV, although IL-7-primed CD4 T cells have a more prominent role. Collectively, we propose intranasal IL-7-mFc pretreatment as an effective means for generating protective immunity against IAV infections, which could be applied to a potential prophylaxis for influenza pandemics in the future. IMPORTANCE: The major consequence of a highly pathogenic IAV infection is severe pulmonary inflammation, which can result in organ failure and death at worst. Although vaccines for seasonal IAVs are effective, frequent variation of surface viral proteins hampers development of protective immunity. In this study, we demonstrated that intranasal IL-7-mFc pretreatment protected immunologically naive mice from lethal IAV infections. Intranasal pretreatment with IL-7-mFc induced an infiltration of T cells in the lung, which reside as effector/memory T cells with lung-retentive markers. Those IL-7-primed pulmonary T cells contributed to development of protective immunity upon IAV infection, reducing pulmonary immunopathology while increasing IAV-specific cytotoxic T lymphocytes. Since a single treatment with IL-7-mFc was effective in the protection against multiple strains of IAV for an extended period of time, our findings suggest a possibility that IL-7-mFc treatment, as a potential prophylaxis, can be developed for controlling highly pathogenic IAV infections.


Subject(s)
Immunoglobulin Fc Fragments/administration & dosage , Immunologic Factors/administration & dosage , Influenza A virus/immunology , Interleukin-7/administration & dosage , Lung/immunology , Orthomyxoviridae Infections/prevention & control , Administration, Intranasal , Animals , Female , Immunoglobulin Fc Fragments/genetics , Immunologic Factors/genetics , Interleukin-7/genetics , Lymphocyte Depletion , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , T-Lymphocytes/immunology
6.
J Immunol ; 195(7): 3001-10, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324771

ABSTRACT

IL-23 is the key cytokine that induces the expansion of Th17 cells. It is composed of p19 and p40 subunits of IL-12. The p40 subunit binds competitively to the receptor of IL-23 and blocks its activity. Our aim was to assess the preventive and therapeutic effect of the IL-12p40 homodimer (p40)2 subunit in autoimmune arthritis animal models. In the current study, using IL-1R antagonist-knockout mice and a collagen-induced arthritis model, we investigated the suppressive effect of (p40)2 on inflammatory arthritis. We demonstrated that the recombinant adenovirus-expressing mouse (p40)2 model prevented the development of arthritis when given before the onset of arthritis. It also decreased the arthritis index and joint erosions in the mouse model if transferred after arthritis was established. (p40)2 inhibited the production of inflammatory cytokines and Ag-specific T cell proliferation. It also induced CD4(+)CD25(+)Foxp3 regulatory T (Treg) cells in vitro and in vivo, whereas the generation of retinoic acid receptor-related organ receptor γt and Th17 cells was suppressed. The induction of Treg cells and the suppression of Th17 cells were mediated via activated STAT5 and suppressed STAT3. Our data suggest that (p40)2 suppressed inflammatory arthritis successfully. This could be a useful therapeutic approach in autoimmune arthritis to regulate the Th17/Treg balance and IL-23 signaling.


Subject(s)
Arthritis, Experimental/prevention & control , Interleukin-12 Subunit p40/pharmacology , Interleukin-23 Subunit p19/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Cell Proliferation/drug effects , Cells, Cultured , Collagen/immunology , Cytokines/biosynthesis , Interleukin-12 Subunit p40/immunology , Interleukin-17/biosynthesis , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred DBA , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/biosynthesis , Protein Multimerization , Receptors, Interleukin/immunology , Receptors, Interleukin-1 Type I/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/immunology
7.
Gut ; 64(2): 260-71, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24902766

ABSTRACT

OBJECTIVE: Programmed death-ligand 1 (PD-L1) has been shown to negatively regulate immune responses via its interaction with PD-1 receptor. In this study, we investigated the effects of PD-L1-Fc treatment on intestinal inflammation using two murine models of inflammatory colitis induced by dextran sulfate sodium (DSS) and T-cell transfer. DESIGN: The anti-colitis effect of adenovirus expressing Fc-conjugated PD-L1 (Ad/PD-L1-Fc) and recombinant PD-L1-Fc protein was evaluated in DSS-treated wild-type and Rag-1 knockout (KO) mice. We examined differentiation of T-helper cells, frequency of innate immune cells, and cytokine production by dendritic cells (DCs) in the colon from DSS-treated mice after PD-L1-Fc administration. In Rag-1 KO mice reconstituted with CD4 CD45RB(high) T cells, we assessed the treatment effect of PD-L1-Fc protein on the development of colitis. RESULTS: Administration of Ad/PD-L1-Fc significantly ameliorated DSS-induced colitis, which was accompanied by diminished frequency of interleukin (IL)-17A-producing CD4 T cells and increased interferon-γ-producing CD4 T cells in the colon of DSS-fed mice. The anti-colitic effect of PD-L1-Fc treatment was also observed in DSS-treated Rag-1 KO mice, indicating lymphoid cell independency. PD-L1-Fc modulated cytokine production by colonic DCs and the effect was dependent on PD-1 expression. Furthermore, PD-L1-Fc protein could significantly reduce the severity of colitis in CD4 CD45RB(high) T-cell-transferred Rag-1 KO mice. CONCLUSIONS: Based on the protective effect of PD-L1-Fc against DSS-induced and T-cell-induced colitis, our results suggest that PD-1-mediated inhibitory signals have a crucial role in limiting the development of colonic inflammation. This implicates that PD-L1-Fc may provide a novel therapeutic approach to treat inflammatory bowel disease.


Subject(s)
B7-H1 Antigen/therapeutic use , Colitis, Ulcerative/prevention & control , Immunologic Factors/therapeutic use , Acute Disease , Adenoviridae/genetics , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , B7-H1 Antigen/pharmacology , Cell Differentiation/drug effects , Colitis, Ulcerative/etiology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/immunology , Cytokines/biosynthesis , Dendritic Cells/immunology , Dextran Sulfate , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Genetic Vectors , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunity, Innate , Immunity, Mucosal , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Fc Fragments/therapeutic use , Immunologic Factors/genetics , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Intestinal Mucosa/immunology , Lymphocyte Transfusion , Mice, Inbred C57BL , Mice, Knockout , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes, Helper-Inducer/immunology , Th17 Cells/immunology
8.
J Leukoc Biol ; 95(4): 621-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24319287

ABSTRACT

PD-1 is a well-established negative regulator of T cell responses by inhibiting proliferation and cytokine production of T cells via interaction with its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), expressed on non-T cells. Recently, PD-1 was found to be expressed in innate cells, including activated DCs, and plays roles in suppressing production of inflammatory cytokines. In this study, we demonstrate that PD-1 KO DCs exhibited prolonged longevity compared with WT DCs in the dLNs after transfer of DCs into hind footpads. Interestingly, upon LPS stimulation, WT DCs increased the expression of PD-1 and started to undergo apoptosis. DCs, in spleen of LPS-injected PD-1 KO mice, were more resistant to LPS-mediated apoptosis in vivo than WT controls. Moreover, treatment of blocking anti-PD-1 mAb during DC maturation resulted in enhanced DC survival, suggesting that PD-1:PD-L interactions are involved in DC apoptosis. As a result, PD-1-deficient DCs augmented T cell responses in terms of antigen-specific IFN-γ production and proliferation of CD4 and CD8 T cells to a greater degree than WT DCs. Moreover, PD-1 KO DCs exhibited increased MAPK1 and CD40-CD40L signaling, suggesting a possible mechanism for enhanced DC survival in the absence of PD-1 expression. Taken together, our findings further extend the function of PD-1, which plays an important role in apoptosis of activated DCs and provides important implications for PD-1-mediated immune regulation.


Subject(s)
Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/physiology , Adoptive Transfer , Animals , Apoptosis , CD40 Antigens/physiology , CD40 Ligand/physiology , Cell Survival , Cells, Cultured , Dendritic Cells/cytology , Lipopolysaccharides/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/physiology , T-Lymphocytes/immunology
9.
Exp Mol Med ; 45: e46, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-24091748

ABSTRACT

Interleukin (IL)-27 is a novel cytokine of the IL-6/IL-12 family that has been reported to be involved in the pathogenesis of autoimmune diseases and has a pivotal role as both a pro- and anti-inflammatory cytokine. We investigated the in vivo effects of IL-27 on arthritis severity in a murine collagen-induced arthritis (CIA) model and its mechanism of action regarding control of regulatory T (Tregs) and IL-17-producing T helper 17 (Th17) cells. IL-27-Fc-treated CIA mice showed a lower severity of arthritis. IL-17 expression in the spleens was significantly decreased in IL-27-Fc-treated CIA mice compared with that in the CIA model. The Th17 population was decreased in the spleens of IL-27-Fc-treated CIA mice, whereas the CD4(+)CD25(+)Foxp3(+) Treg population increased. In vitro studies revealed that IL-27 inhibited IL-17 production in murine CD4(+) T cells, and the effect was associated with retinoic acid-related orphan receptor γT and signal transducer and activator of transcription 3 inhibition. In contrast, fluorescein isothiocyanate-labeled forkhead box P3 (Foxp3) and IL-10 were profoundly augmented by IL-27 treatment. Regarding the suppressive capacity of Treg cells, the proportions of CTLA-4(+) (cytotoxic T-lymphocyte antigen 4), PD-1(+) (programmed cell death protein 1) and GITR(+) (glucocorticoid-induced tumor necrosis factor receptor) Tregs increased in the spleens of IL-27-Fc-treated CIA mice. Furthermore, in vitro differentiated Treg cells with IL-27 exerted a more suppressive capacity on T-cell proliferation. We found that IL-27 acts as a reciprocal regulator of the Th17 and Treg populations in CD4(+) cells isolated from healthy human peripheral blood mononuclear cells (PBMCs), as well as from humans with rheumatoid arthritis (RA) PBMCs. Our study suggests that IL-27 has the potential to ameliorate overwhelming inflammation in patients with RA through a reciprocal regulation of Th17 and Treg cells.


Subject(s)
Arthritis, Experimental/drug therapy , Interleukins/therapeutic use , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/immunology , Cells, Cultured , Humans , Interleukins/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA
10.
Vaccine ; 31(27): 2884-90, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23624092

ABSTRACT

Improvement to the immunogenicity of DNA vaccines was evaluated in a Mycobacterium tuberculosis (MTB) infection mouse model examining the combined effects of nonlytic Fc-fused IL-7 DNA (IL-7-nFc) and Flt3-ligand fused Mtb32 (F-Mtb32) DNA. Mice were treated with conventional chemotherapy for 6 weeks from 4 weeks after aerosol infection of MTB. Following the start of chemotherapy, DNA immunizations were administered five times with 2-week intervals. Coadministration of IL-7-nFc and F-Mtb32 DNA given during chemotherapy synergistically enhanced the magnitude of Mtb32-specific T cell responses and sustained for one-year after the last immunization assessed by IFN-γ ELISPOT assay. After dexamethasone treatment, a significantly reduced MTB reactivation was observed in mice received both IL-7-nFc and F-Mtb32 DNA, compared with F-MTb32 DNA alone or with control mice. In addition, mice treated with IL-7-nFc and F-Mtb32 DNA together showed improved lung pathology and reduced pulmonary inflammation values relative to F-Mtb32 DNA or saline injected mice. Intracellular cytokine staining revealed that the protection levels induced by combination therapy with IL-7-nFc and F-Mtb32 DNA was associated with enhanced Mtb32-specific IFN-γ secreting CD4(+) T cell responses and CD8(+) T cell responses stimulated with CTL epitope peptide in the lungs and spleens. These data suggest that IL-7-nFc as a novel TB adjuvant may facilitate therapeutic TB DNA vaccine to the clinics through significant enhancement of codelivered DNA vaccine-induced T cell immunity.


Subject(s)
Antigens, Bacterial/immunology , Immunoglobulin Fc Fragments/immunology , Interleukin-7/immunology , Membrane Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, DNA/immunology , Animals , Antigens, Bacterial/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Synergism , Female , Immunoglobulin Fc Fragments/genetics , Interferon-gamma/biosynthesis , Interleukin-7/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Vaccination
11.
Immunology ; 137(4): 326-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22812379

ABSTRACT

Interleukin (IL)-27 is a heterodimeric cytokine that is known to have both stimulatory and inhibitory functions during immune responses. We investigated the effects of IL-27 on arthritis and bone erosion in the murine collagen-induced arthritis (CIA) model. We demonstrate that the inhibitory effect of IL-27 on osteoclastogenesis is associated with interferon-γ (IFN-γ) production by using an IFN-γ knockout mouse model. The IL-27-Fc was injected into both CIA and IFN-γ-deficient mice. The effects of IL-27-Fc on osteoclast differentiation were evaluated both in vitro and in vivo. The IL-27-Fc-injected mice showed significantly lower arthritis indices and fewer tartrate-resistant acid-phosphatase-positive osteoclasts in their joint tissues than untreated mice. Interleukin-27 inhibited osteoclastogenesis from bone marrow-derived mononuclear cells in vitro, which was counteracted by the addition of anti-IFN-γ antibody. The IL-27-Fc did not affect arthritis in IFN-γ knockout mice. Interleukin-27 also suppressed osteoclast differentiation in human and intriguingly, it could promote the expression of IFN-γ on priming osteoclasts. These results imply that IL-27 suppressed the generation of CIA and osteoclastogenesis, which were mediated by the induction of IFN-γ.


Subject(s)
Interferon-gamma/biosynthesis , Interleukin-17/pharmacology , Osteoclasts/drug effects , Osteogenesis/drug effects , Animals , Arthritis, Experimental/etiology , Cell Differentiation/drug effects , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Osteoclasts/physiology
SELECTION OF CITATIONS
SEARCH DETAIL