Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 5382, 2023 09 04.
Article in English | MEDLINE | ID: mdl-37666819

ABSTRACT

Regulatory T cells (Treg) are CD4+ T cells with immune-suppressive function, which is defined by Foxp3 expression. However, the molecular determinants defining the suppressive population of T cells have yet to be discovered. Here we report that the cell surface protein Lrig1 is enriched in suppressive T cells and controls their suppressive behaviors. Within CD4+ T cells, Treg cells express the highest levels of Lrig1, and the expression level is further increasing with activation. The Lrig1+ subpopulation from T helper (Th) 17 cells showed higher suppressive activity than the Lrig1- subpopulation. Lrig1-deficiency impairs the suppressive function of Treg cells, while Lrig1-deficient naïve T cells normally differentiate into other T cell subsets. Adoptive transfer of CD4+Lrig1+ T cells alleviates autoimmune symptoms in colitis and lupus nephritis mouse models. A monoclonal anti-Lrig1 antibody significantly improves the symptoms of experimental autoimmune encephalomyelitis. In conclusion, Lrig1 is an important regulator of suppressive T cell function and an exploitable target for treating autoimmune conditions.


Subject(s)
Autoimmunity , Colitis , Animals , Mice , CD4-Positive T-Lymphocytes , T-Lymphocytes, Regulatory , Adoptive Transfer , Transcription Factors , Forkhead Transcription Factors/genetics
2.
Biochem Biophys Res Commun ; 640: 32-39, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36502629

ABSTRACT

Although the T helper 2 (Th2) subset is a critical player in the humoral immune response to extracellular parasites and suppression of Th1-mediated inflammation, Th2 cells have been implicated in allergic inflammatory diseases such as asthma, allergic rhinitis, and atopic dermatitis. GATA binding protein 3 (GATA3) is a primary transcription factor that mediates Th2 differentiation and secretion of Th2 cytokines, including IL-4, IL-5, and IL-13. Here, a nucleus-deliverable form of GATA3-transcription modulation domain (TMD) (ndG3-TMD) was generated using Hph-1 human protein transduction domain (PTD) to modulate the transcriptional function of endogenous GATA3 without genetic manipulation. ndG3-TMD was shown to be efficiently delivered into the cell nucleus quickly without affecting cell viability or intracellular signaling events for T cell activation. ndG3-TMD exhibited a specific inhibitory function for the endogenous GATA3-mediated transcription, such as Th2 cell differentiation and Th2-type cytokine production. Intranasal administration of ndG3-TMD significantly alleviated airway hyperresponsiveness, infiltration of immune cells, and serum IgE level in an OVA-induced mouse model of asthma. Also, Th2 cytokine secretion by the splenocytes isolated from the ndG3-TMD-treated mice substantially decreased. Our results suggest that ndG3-TMD can be a new therapeutic reagent to suppress Th2-mediated allergic diseases through intranasal delivery.


Subject(s)
Asthma , GATA3 Transcription Factor , Respiratory Hypersensitivity , Animals , Humans , Mice , Administration, Intranasal , Asthma/therapy , Cell Nucleus/metabolism , Cytokines/metabolism , Disease Models, Animal , GATA3 Transcription Factor/administration & dosage , GATA3 Transcription Factor/chemistry , Mice, Inbred BALB C , Ovalbumin , Respiratory Hypersensitivity/therapy , Th2 Cells
3.
Front Immunol ; 12: 741938, 2021.
Article in English | MEDLINE | ID: mdl-34745114

ABSTRACT

T helper 17 (TH17) cells are involved in several autoimmune diseases such as multiple sclerosis (MS) and rheumatoid arthritis (RA). In addition to retinoic acid receptor-related orphan nuclear receptor gamma t (ROR-γt), hypoxia-inducible factor-1α (HIF-1α) is essential for the differentiation and inflammatory function of TH17 cells. To investigate the roles of HIF-1α in the functional regulation of TH17 cells under the normal physiological condition without genetic modification, the nucleus-transducible form of transcription modulation domain (TMD) of HIF-1α (ntHIF-1α-TMD) was generated by conjugating HIF-1α-TMD to Hph-1 protein transduction domain (PTD). ntHIF-1α-TMD was effectively delivered into the nucleus of T cells without cellular cytotoxicity. ntHIF-1α-TMD significantly blocked the differentiation of naïve T cells into TH17 cells in a dose-dependent manner via IL-17A and ROR-γt expression inhibition. However, T-cell activation events such as induction of CD69, CD25, and IL-2 and the differentiation potential of naïve T cells into TH1, TH2, or Treg cells were not affected by ntHIF-1α-TMD. Interestingly, TH17 cells differentiated from naïve T cells in the presence of ntHIF-1α-TMD showed a substantial level of suppressive activity toward the activated T cells, and the increase of Foxp3 and IL-10 expression was detected in these TH17 cells. When mRNA expression pattern was compared between TH17 cells and ntHIF-1α-TMD-treated TH17 cells, the expression of the genes involved in the differentiation and functions of TH17 cells was downregulated, and that of the genes necessary for immune-suppressive functions of Treg cells was upregulated. When the mice with experimental autoimmune encephalomyelitis (EAE) were treated with ntHIF-1α-TMD with anti-IL-17A mAb as a positive control, the therapeutic efficacy of ntHIF-1α-TMD in vivo was comparable with that of anti-IL-17A mAb, and ntHIF-1α-TMD-mediated therapeutic effect was contributed by the functional conversion of TH17 cells into immune-suppressive T cells. The results in this study demonstrate that ntHIF-1α-TMD can be a new therapeutic reagent for the treatment of various autoimmune diseases in which TH17 cells are dominant and pathogenic T cells.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Female , Mice , Mice, Inbred C57BL
4.
Inflammation ; 44(6): 2291-2301, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34169410

ABSTRACT

The objective of this study was to find out if suppression of NF-kB complex function by p65-TMD-linked PTD could reduce host inflammation and bone resorption at peri-implantitis sites in rats. Twenty-one male 5-week-old SD rats were divided into three groups: untreated control group (A), silk-induced peri-implantitis group (B), and nt (nucleus transducible)-p65-TMD-treated, silk-induced peri-implantitis group (C). Implant sulcus of a rat in group C were divided into two groups, namely group Cp and Cb. Palatal implant sulcus where nt-p65-TMD solution was applied with an insulin syringe were assigned to group Cp. Buccal implant sulcus without topical nt-p65-TMD application were assigned to group Cb. H&E staining, TRAP staining, and immunohistological staining were done. The crestal bone levels of group A were significantly higher than those of group B at p<0.01. The crestal bone levels of group Cp were significantly higher than those of group Cb at p<0.05. H-E staining showed increased apical migration of junctional epithelium and inflammatory cells in group Cb. TRAP staining revealed more multinucleated osteoclasts in group Cb. As for immunohistological staining, group Cb showed many IL-6-positive cells while group Cp had none. In this study, p65-TMD-linked PTD inhibited NF-kB functions and reduced inflammation and bone resorption at peri-implantitis sites in rats.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bone Resorption/prevention & control , Inflammation Mediators/antagonists & inhibitors , Inflammation/prevention & control , Jaw/drug effects , NF-kappa B/antagonists & inhibitors , Peri-Implantitis/prevention & control , Animals , Bone Resorption/immunology , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Screws , Bone-Implant Interface/pathology , Disease Models, Animal , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Jaw/immunology , Jaw/metabolism , Jaw/pathology , Male , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoclasts/pathology , Peri-Implantitis/immunology , Peri-Implantitis/metabolism , Peri-Implantitis/pathology , Rats, Sprague-Dawley
5.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671455

ABSTRACT

After avulsion and replantation, teeth are at risk of bone and root resorption. The present study aimed to demonstrate that the intra-nuclear transducible form of transcription modulation domain of p65 (nt-p65-TMD) can suppress osteoclast differentiation in vitro, and reduce bone resorption in a rat model of tooth replantation. Cell viability and nitric oxide release were evaluated in RAW264.7 cells using CCK-8 assay and Griess reaction kit. Osteoclast differentiation was evaluated using quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and tartrate-resistant acid phosphatase (TRAP) staining. Thirty-two maxillary rat molars were extracted and stored in saline (n = 10) or 10 µM nt-p65-TMD solution (n = 22) before replantation. After 4 weeks, specimens were scored according to the inflammatory pattern using micro-computed tomography (CT) imaging and histological analyses. nt-p65-TMD treatment resulted in significant reduction of nitric oxide release and osteoclast differentiation as studied using PCR and TRAP staining. Further, micro-CT analysis revealed a significant decrease in bone resorption in the nt-p65-TMD treatment group (p < 0.05). Histological analysis of nt-p65-TMD treatment group showed that not only bone and root resorption, but also inflammation of the periodontal ligament and epithelial insertion was significantly reduced. These findings suggest that nt-p65-TMD has the unique capabilities of regulating bone remodeling after tooth replantation.


Subject(s)
Cell Nucleus/metabolism , Tooth Replantation , Transcription Factor RelA/metabolism , Animals , Cell Differentiation , Cell Survival , Mice , Models, Animal , Molar/diagnostic imaging , Nitric Oxide/metabolism , Osteoclasts/metabolism , RAW 264.7 Cells , Rats , Transduction, Genetic , X-Ray Microtomography
6.
Molecules ; 24(4)2019 Feb 20.
Article in English | MEDLINE | ID: mdl-30791543

ABSTRACT

Patients with uncontrolled diabetes are susceptible to implant failure due to impaired bone metabolism. Hypoxia-inducible factor 1α (HIF-1α), a transcription factor that is up-regulated in response to reduced oxygen during bone repair, is known to mediate angiogenesis and osteogenesis. However, its function is inhibited under hyperglycemic conditions in diabetic patients. This study thus evaluates the effects of exogenous HIF-1α on bone formation around implants by applying HIF-1α to diabetic mice and normal mice via a protein transduction domain (PTD)-mediated DNA delivery system. Implants were placed in the both femurs of diabetic and normal mice. HIF-1α and placebo gels were injected to implant sites of the right and left femurs, respectively. We found that bone-to-implant contact (BIC) and bone volume (BV) were significantly greater in the HIF-1α treated group than placebo in diabetic mice (p < 0.05). Bioinformatic analysis showed that diabetic mice had 216 differentially expressed genes (DEGs) and 21 target genes. Among the target genes, NOS2, GPNMB, CCL2, CCL5, CXCL16, and TRIM63 were found to be associated with bone formation. Based on these results, we conclude that local administration of HIF-1α via PTD may boost bone formation around the implant and induce gene expression more favorable to bone formation in diabetic mice.


Subject(s)
DNA-Binding Proteins/metabolism , Dental Implants , Diabetes Mellitus, Experimental , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Osteogenesis/drug effects , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation/drug effects , Immunohistochemistry , Mice , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , Osteogenesis/genetics , Permeability
7.
Int J Hematol ; 108(4): 375-383, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29978433

ABSTRACT

The BAF57 subunit, an indispensable member of the BAF complex, is functionally implicated in apoptosis, cell cycle, and T cell development through chromosomal remodeling. However, the precise roles of BAF57 in the T cell receptor (TcR)-mediated signaling pathway have not been elucidated. In this study, a nucleus-transducible form of BAF57, absent the proline-rich and HMG domains (ntBAF57-ΔPH), was generated to interfere with the interaction between BAF57 and its binding protein, BAF155. ntBAF57-ΔPH was effectively delivered into mouse CD4+ T cells in a dose- and time-dependent manner, without cellular toxicity. Inhibition of T cell activation by ntBAF57-ΔPH was mediated by its disruption of the interaction between BAF155 and BAF57, leading to the degradation of endogenous BAF57 and BAF155. This phenomenon led to alterations in gene expression similar to those associated with Ciclosporin A treatment. In vivo administration of ntBAF57-ΔPH enhanced survival rate of sepsis-induced mice and reduced the LPS-induced secretion of pro-inflammatory cytokines and the expression of endogenous BAF57. These results reveal a novel function of BAF57 as an essential regulator of T cell activation. ntBAF57-ΔPH represents a novel immune-suppressive drug candidate with potential uses in the treatment of autoimmunity and graft rejection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Chromosomal Proteins, Non-Histone/immunology , Immunosuppression Therapy , Lymphocyte Activation , Animals , CD4-Positive T-Lymphocytes/pathology , Chromosomal Proteins, Non-Histone/genetics , Cyclosporine/pharmacology , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred BALB C , Sepsis/drug therapy , Sepsis/genetics , Sepsis/immunology , Sepsis/pathology , Transcription Factors/genetics , Transcription Factors/immunology
8.
Kidney Int ; 93(5): 1118-1130, 2018 05.
Article in English | MEDLINE | ID: mdl-29409726

ABSTRACT

Excessive expression of Tbet and IFNγ is evidence of systemic lupus erythematosus (SLE) in lupus patients. In this study, the nucleus-transducible form of Transcription Modulation Domain (TMD) of Tbet (ntTbet-TMD), which is a fusion protein between Protein Transduction Domain Hph-1 (Hph-1-PTD) and the TMD of Tbet comprising DNA binding domain and isotype-specific domain, was generated to inhibit Tbet-mediated transcription in the interactomic manner. ntTbet-TMD was effectively delivered into the nucleus of the cells and specifically inhibited Tbet-mediated transcription without influencing the differentiation of other T cell subsets and signaling events for T cell activation. The severity of nephritis was significantly reduced by ntTbet-TMD as effectively as methylprednisolone in lupus-prone mice. The number of Th1, Th2 or Th17 cells and the secretion of their cytokines substantially decreased in the spleen and kidney of lupus-prone mice by ntTbet-TMD treatment. In contrast to methylprednisolone, the marked increase of Treg cells and the secretion of their immunosuppressive cytokine were detected in the spleen of (NZB/NZW) F1 mice treated with ntTbet-TMD. Thus, ntTbet-TMD can improve nephritis in lupus-prone mice by modulating the overall proinflammatory microenvironment and rebalancing T cell subsets, leading to new immune therapeutics for Th1-mediated autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Nucleus/drug effects , Kidney/drug effects , Lupus Nephritis/drug therapy , T-Box Domain Proteins/pharmacology , Transcription, Genetic/drug effects , Active Transport, Cell Nucleus , Animals , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cellular Microenvironment , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Mice, Inbred NZB , Protein Domains , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
9.
Mol Cell Biochem ; 437(1-2): 99-107, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28660411

ABSTRACT

Hypoxia-inducible factor-1 alpha (HIF1A) is an important transcription factor for angiogenesis. Recent studies have used the protein transduction domain (PTD) to deliver genes, but the PTD has not been used to induce the expression of HIF1A. This study aimed at using a novel PTD (Hph-1-GAL4; ARVRRRGPRR) to overexpress the HIF1A and identify the effects on angiogenesis in vitro and in vivo. Overexpression of HIF1A was induced using Hph-1-GAL4 in human umbilical vein/vascular endothelium cells (HUVEC). The expression levels of genes were analyzed by the quantitative real-time polymerase chain reaction (qPCR) after 2 and 4 days, respectively. An in vitro tube formation was performed using Diff-Quik staining. HIF1A and Hph-1-GAL4 were injected subcutaneously into the ventral area of each 5-week-old mouse. All of the plugs were retrieved after 1 week, and the gene expression levels were evaluated by qPCR. Each Matrigel plug was evaluated using the hemoglobin assay and hematoxylin and eosin (HE) staining. The expression levels of HIF1A and HIF1A target genes were significantly higher in HIF1A-transfected HUVEC than in control HUVEC in vitro. In the in vivo Matrigel plug assay, the amount of hemoglobin was significantly higher in the HIF1A-treatment group than in the PBS-treatment group. Blood vessels were identified in the HIF1A-treatment group. The expression levels of HIF1A, vascular endothelial growth factor (Vegf), and Cd31 were significantly higher in the HIF1A-treatment group than in the PBS-treatment group. These findings suggest that using Hph-1-G4D to overexpress HIF1A might be useful for transferring genes and regenerating tissues.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Neovascularization, Physiologic/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Humans , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
10.
Arch Plast Surg ; 44(1): 76-79, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28194352

ABSTRACT

Necrotizing fasciitis (NF) is an aggressive soft-tissue infection involving the deep fascia and is characterized by extensive deterioration of the surrounding tissue. Immediate diagnosis and intensive treatment, including debridement and systemic antibiotics, represent the most important factors influencing the survival of NF patients. In this report, we present a case of NF in the abdomen due to an infection caused by a perforated small bowel after abdominal liposuction. It was successfully treated using negative-pressure wound therapy, in which a silicone sheet functioned as a barrier between the sponge and internal organs to protect the small bowel.

SELECTION OF CITATIONS
SEARCH DETAIL
...