Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 32(2): 101256, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38774582

ABSTRACT

Glycosylation of biopharmaceuticals can affect their safety and efficacy. Glycans can occur on recombinant adeno-associated viruses (rAAVs) that are used for gene therapy; however, the types of glycans that attach to rAAVs are controversial. Here, we conducted lectin microarray analyses on six rAAV serotype 6 (rAAV6) preparations that were produced differently. We demonstrate that O-glycans considered to be attached to rAAV6 were recognized by Agaricus bisporus agglutinin (ABA) and that N-glycans were detected in rAAV6 purified without affinity chromatography. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis showed that the N-glycans detected in rAAV6 were derived from host cell proteins. A combination of ABA-based fractionation and LC-MS/MS revealed that rAAV6 was O-glycosylated with the mucin-type glycans, O-GalNAc (Tn antigen), and mono- and di-sialylated Galß1-3GalNAc (T antigen) at S156, T162, T194, and T201 in viral protein (VP) 2 and with O-GlcNAc at T242 in VP3. The mucin-type O-glycosylated rAAV6 particles were 0.1%-1% of total particles. Further physicochemical and biological analyses revealed that mucin-type O-glycosylated rAAV6 had a lower ratio of VP1 to VP2/VP3, resulting in a lower transduction efficiency both in vitro and in vivo compared with rAAV6 without mucin-type O-glycans. This report details conclusive evidence of rAAV glycosylation and its impact on rAAV-based therapeutics.

2.
Blood Adv ; 2024 May 31.
Article in English | MEDLINE | ID: mdl-38820442

ABSTRACT

The A2 domain dissociation in activated factor (F)VIII (FVIIIa) results in reduced activity. Previous studies demonstrated that some FVIII mutants (D519V/E665V and K1813A) with delayed A2 dissociation enhanced coagulation potential. We speculated, therefore, that FVIII encompassing a combination of these mutations might further enhance coagulant activity. Aim is to assess the D519V/E665V/K1813A-FVIII mutation as a gain-of-function. The FVIII mutants, D519V/E665V/K1813A, D519V/E665V, and K1813A were expressed in a BHK cell system, and global coagulation potential of these mutants was compared with WT FVIII in vitro and in hemophilia A mice in vivo. Kinetic analyses indicated that the apparent Kd for FIXa on the tenase assembly with D519V/E665V and D519V/E665V/K1813A mutants were lower, and that the generated FXa for D519V/E665V/K1813A was significantly greater than WT. The WT-FVIII activity after thrombin activation increased by ~12-fold within 5 minutes, and returned to initial levels within 30 minutes. In contrast, The FVIII-related activity of D519V/E665V/K1813A increased further with time after thrombin activation, and showed an ~25-fold increase at 2 hours. The A2 dissociation rate of D519V/E665V/K1813A was ~50-fold slower than WT in a one-stage clotting assay. Thrombin generation assays demonstrated that D519V/E665V/K1813A (0.125 nM) exhibited coagulation potential comparable to WT (1 nM). In animal studies, rotational thromboelastometry and tail-clip assays showed that the coagulation potential of D519V/E665V/K1813A (0.25 µg/kg) was equal to WT (2 µg/kg). FVIII-D519V/E665V/K1813A mutant could provide an ~8-fold increase in hemostatic function of WT FVIII, due to increased FVIIIa stability and the association between FVIIIa and FIXa.

3.
Blood Adv ; 7(22): 7017-7027, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37792826

ABSTRACT

The importance of genetic diagnosis for patients with hemophilia has been recently demonstrated. However, the pathological variant cannot be identified in some patients. Here, we aimed to identify the pathogenic intronic variant causing hemophilia A using induced pluripotent stem cells (iPSCs) from patients and genome editing. We analyzed siblings with moderate hemophilia A and without abnormalities in the F8 exon. Next-generation sequencing of the entire F8 revealed 23 common intron variants. Variant effect predictor software indicated that the deep intronic variant at c.5220-8563A>G (intron 14) might act as a splicing acceptor. We developed iPSCs from patients and used genome editing to insert the elongation factor 1α promoter to express F8 messenger RNA (mRNA). Then, we confirmed the existence of abnormal F8 mRNA derived from aberrant splicing, resulting in a premature terminal codon as well as a significant reduction in F8 mRNA in iPSCs due to nonsense-mediated RNA decay. Gene repair by genome editing recovered whole F8 mRNA expression. Introduction of the intron variant into human B-domain-deleted F8 complementary DNA suppressed factor VIII (FVIII) activity and produced abnormal FVIII lacking the light chain in HEK293 cells. Furthermore, genome editing of the intron variant restored FVIII production. In summary, we have directly proven that the deep intronic variant in F8 results in aberrant splicing, leading to abnormal mRNA and nonsense-mediated RNA decay. Additionally, genome editing targeting the variant restored F8 mRNA and FVIII production. Our approach could be useful not only for identifying causal variants but also for verifying the therapeutic effect of personalized genome editing.


Subject(s)
Hemophilia A , Hemostatics , Induced Pluripotent Stem Cells , Humans , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia A/diagnosis , Gene Editing , Induced Pluripotent Stem Cells/metabolism , HEK293 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Mol Ther Methods Clin Dev ; 30: 502-514, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37693948

ABSTRACT

Gene therapy using adeno-associated virus (AAV)-based vectors has become a realistic therapeutic option for hemophilia. We examined the potential of a novel engineered liver-tropic AAV3B-based vector, AAV.GT5, for hemophilia B gene therapy. In vitro transduction with AAV.GT5 in human hepatocytes was more than 100 times higher than with AAV-Spark100, another bioengineered vector used in a clinical trial. However, liver transduction following intravenous injection of these vectors was similar in mice with a humanized liver and in macaques. This discrepancy was due to the low recovery and short half-life of AAV.GT5 in blood, depending on the positive charge of the heparin-binding site in the capsid. Bypassing systemic clearance with the intra-hepatic vascular administration of AAV.GT5, but not AAV-Spark100, enhanced liver transduction in pigs and macaques. AAV.GT5 did not develop neutralizing antibodies (NAbs) in two of four animals, while AAV-Spark100 induced serotype-specific NAbs in all macaques tested (4 of 4). The NAbs produced after AAV-Spark100 administration were relatively serotype specific, and challenge with AAV.GT5 through the hepatic artery successfully boosted liver transduction in one animal previously administered AAV-Spark100. In summary, AAV.GT5 showed different vector kinetics and NAb induction compared with AAV-Spark100, and intra-hepatic vascular administration may minimize the vector dose required and vector dissemination.

5.
Rinsho Ketsueki ; 64(5): 377-388, 2023.
Article in Japanese | MEDLINE | ID: mdl-37271529

ABSTRACT

Hemophiliacs are X-linked inherited bleeding disorders. The treatment of hemophilia is based on periodic replacement treatment with coagulation factor products to avoid the development of hemophilic arthropathy, as well as to control fatal bleeding. However, because of the coagulation factor's incredibly short half-life, the medication must be often given intravenously throughout one's life. Extended half-life coagulation factor preparations have lowered the burden of treatment by enhancing pharmacokinetics. However, the patients remain at risk of developing inhibitors to significantly interfere with the therapeutic effectiveness of coagulation factor concentrates. Recently, nonfactor replacement therapies have gained a lot of interest as a new class of therapeutic drugs preventing bleeding in patients with or without inhibitors. Moreover, gene therapy drugs for hemophilia have recently been approved in Europe and the United States. Because gene therapy can preserve coagulation factor levels in the blood for at least several years with a single treatment, it may become a curative treatment that removes the need for the therapy for a long time.


Subject(s)
Hematologic Diseases , Hemophilia A , Humans , Hemophilia A/drug therapy , Blood Coagulation Factors/therapeutic use , Blood Coagulation , Hematologic Diseases/therapy , Genetic Therapy , Factor VIII/therapeutic use
6.
Commun Med (Lond) ; 3(1): 56, 2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37076593

ABSTRACT

BACKGROUND: Base editing via CRISPR-Cas9 has garnered attention as a method for correcting disease-specific mutations without causing double-strand breaks, thereby avoiding large deletions and translocations in the host chromosome. However, its reliance on the protospacer adjacent motif (PAM) can limit its use. We aimed to restore a disease mutation in a patient with severe hemophilia B using base editing with SpCas9-NG, a modified Cas9 with the board PAM flexibility. METHODS: We generated induced pluripotent stem cells (iPSCs) from a patient with hemophilia B (c.947T>C; I316T) and established HEK293 cells and knock-in mice expressing the patient's F9 cDNA. We transduced the cytidine base editor (C>T), including the nickase version of Cas9 (wild-type SpCas9 or SpCas9-NG), into the HEK293 cells and knock-in mice through plasmid transfection and an adeno-associated virus vector, respectively. RESULTS: Here we demonstrate the broad PAM flexibility of SpCas9-NG near the mutation site. The base-editing approach using SpCas9-NG but not wild-type SpCas9 successfully converts C to T at the mutation in the iPSCs. Gene-corrected iPSCs differentiate into hepatocyte-like cells in vitro and express substantial levels of F9 mRNA after subrenal capsule transplantation into immunodeficient mice. Additionally, SpCas9-NG-mediated base editing corrects the mutation in both HEK293 cells and knock-in mice, thereby restoring the production of the coagulation factor. CONCLUSION: A base-editing approach utilizing the broad PAM flexibility of SpCas9-NG can provide a solution for the treatment of genetic diseases, including hemophilia B.


In patients with hemophilia B, the blood does not clot properly, leading to excessive bruising and bleeding. Hemophilia B is caused by an error in a gene called coagulation factor IX (F9). To treat patients with hemophilia B, we might be able to use a technology called CRISPR-Cas9 to edit and correct this genetic error, restoring factor IX function and improving clotting. Here, we test a specific CRISPR-Cas9 approach in cells and animals. We show that we are able to correct the genetic error in F9 in cells isolated from a patient with severe hemophilia B. We also show that we can fix the error in mice and that this increases levels of factor IX in the blood of the mice. With further testing, this gene-editing approach may be a viable therapy for patients with hemophilia B or similar genetic disorders.

7.
J Gene Med ; 25(8): e3505, 2023 08.
Article in English | MEDLINE | ID: mdl-36972408

ABSTRACT

BACKGROUND: Intravenous administration of adeno-associated virus (AAV) vectors is a promising gene therapy approach for monogenic diseases. However, re-administration of the same AAV serotype is impossible because of the induction of anti-AAV neutralizing antibodies (NAbs). Here, we examined the feasibility of re-administrating AAV vector serotypes different from the initial AAV vector serotype. METHODS: Liver-targeting AAV3B, AAV5, and AAV8 vectors were intravenously injected in C57BL/6 mice, and the emergence of NAbs and the transduction efficacy following re-administration were evaluated. RESULTS: For all serotypes, re-administration of the same serotype was not possible. Although the highest neutralizing activity of NAb was induced by AAV5, anti-AAV5 NAbs did not react with other serotypes, resulting in successful re-administration with the other serotypes. AAV5 re-administration was also successful in all mice treated with AAV3B and AAV8. Effective secondary administration of AAV3B and AAV8 was observed in most mice initially administrated AAV8 and AAV3B, respectively. However, few mice developed NAbs cross-reacting with the other serotypes, especially those with close sequence homology. CONCLUSIONS: In summary, AAV vector administration induced NAbs relatively specific to the administrated serotype. Secondary administration of AAVs targeting liver transduction could be successfully achieved by switching AAV serotypes in mice.


Subject(s)
Dependovirus , Genetic Vectors , Animals , Mice , Dependovirus/genetics , Genetic Vectors/genetics , Mice, Inbred C57BL , Liver , Antibodies, Neutralizing
8.
Methods Mol Biol ; 2637: 195-211, 2023.
Article in English | MEDLINE | ID: mdl-36773148

ABSTRACT

Adeno-associated virus (AAV) vectors are attractive tools for gene transfer to the liver and are used as gene therapeutic drugs for inherited disorders. The intravenous injection of an AAV vector harboring the gene of interest driven by the hepatocyte-specific promoter could efficiently express the target gene in liver hepatocytes. The delivery of genome editing tools including Cas9 and gRNA, by the AAV vector, can efficiently disrupt the target gene expression in the liver in vivo by intravenous administration in mice. We can quickly obtain mice lacking specific gene expression in the liver only by administering the AAV vector. The method could be suitable for developing genome editing treatments for inherited disorders and basic research exploring the physiological role of the target gene produced from liver hepatocytes.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Mice , CRISPR-Cas Systems/genetics , Liver/metabolism , Hepatocytes , Promoter Regions, Genetic , Dependovirus/genetics , Genetic Vectors/genetics
9.
Mol Ther Methods Clin Dev ; 27: 404-414, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36381300

ABSTRACT

Adeno-associated virus (AAV) vectors are promising modalities of gene therapy to address unmet medical needs. However, anti-AAV neutralizing antibodies (NAbs) hamper the vector-mediated therapeutic effect. Therefore, NAb prevalence in the target population is vital in designing clinical trials with AAV vectors. Hence, updating the seroprevalence of anti-AAV NAbs, herein we analyzed sera from 100 healthy individuals and 216 hemophiliacs in Japan. In both groups, the overall seroprevalence against various AAV serotypes was 20%-30%, and the ratio of the NAb-positive population increased with age. The seroprevalence did not differ between healthy participants and hemophiliacs and was not biased by the concomitant blood-borne viral infections. The high neutralizing activity, which strongly inhibits the transduction with all serotypes in vitro, was mostly found in people in their 60s or of older age. The multivariate analysis suggested that "60s or older age" was the only independent factor related to the high titer of NAbs. Conversely, a large proportion of younger hemophiliacs was seronegative, rendering them eligible for AAV-mediated gene therapy in Japan. Compared with our previous study, the peak of seroprevalences has shifted to older populations, indicating that natural AAV exposure in the elderly occurred in their youth but not during the last decade.

10.
Mol Ther Methods Clin Dev ; 22: 162-171, 2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34485602

ABSTRACT

Most gene therapy clinical trials that systemically administered adeno-associated virus (AAV) vector enrolled only patients without anti-AAV-neutralizing antibodies. However, laboratory tests to measure neutralizing antibodies varied among clinical trials and have not been standardized. In this study, we attempted to improve the sensitivity and reproducibility of a cell-based assay to detect neutralizing antibodies and to determine the detection threshold to predict treatment efficacy. Application of the secreted type of NanoLuc and AAV receptor-expressing cells reduced the multiplicity of infection (MOI) for AAV transduction and improved the sensitivity to detect neutralizing antibodies with a low coefficient of variation, whereas the detection threshold could not be improved by the reduction of MOI to <100. After human immunoglobulin administration into mice at various doses, treatment with high-dose AAV8 vector enabled evasion of the inhibitory effect of neutralizing antibodies. Conversely, gene transduction was slightly influenced in the mice treated with low-dose AAV8 vector, even when neutralizing antibodies were determined to be negative in the assay. In conclusion, we developed a reliable and sensitive cell-based assay to measure neutralizing antibodies against AAV and found that the appropriate MOI to detect marginal neutralizing antibodies was 100. Other factors, including noninhibitory antibodies, marginally influence in vivo transduction at low vector doses.

11.
Sci Rep ; 11(1): 14824, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34290295

ABSTRACT

Coagulation factors are produced from hepatocytes, whereas production of coagulation factor VIII (FVIII) from primary tissues and cell species is still controversial. Here, we tried to characterize primary FVIII-producing organ and cell species using genetically engineered mice, in which enhanced green fluorescent protein (EGFP) was expressed instead of the F8 gene. EGFP-positive FVIII-producing cells existed only in thin sinusoidal layer of the liver and characterized as CD31high, CD146high, and lymphatic vascular endothelial hyaluronan receptor 1 (Lyve1)+. EGFP-positive cells can be clearly distinguished from lymphatic endothelial cells in the expression profile of the podoplanin- and C-type lectin-like receptor-2 (CLEC-2)+. In embryogenesis, EGFP-positive cells began to emerge at E14.5 and subsequently increased according to liver maturation. Furthermore, plasma FVIII could be abolished by crossing F8 conditional deficient mice with Lyve1-Cre mice. In conclusion, in mice, FVIII is only produced from endothelial cells exhibiting CD31high, CD146high, Lyve1+, CLEC-2+, and podoplanin- in liver sinusoidal endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Factor VIII/biosynthesis , Liver/cytology , Animals , CD146 Antigen/metabolism , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Lectins, C-Type/metabolism , Liver/embryology , Membrane Glycoproteins/metabolism , Mice , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Vesicular Transport Proteins/metabolism
12.
Scand J Immunol ; 93(6): e13020, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33393095

ABSTRACT

CD4+ CD25+ Foxp3+ regulatory T cells (Tregs) are highly sensitive to IL-2, one of the many cytokines produced during immune responses, for their development, survival and functions. Although the effects of IL-2 administration on Tregs in vivo are well characterized, the effects on Tregs elicited by IL-2 produced during an immune response have not been elucidated. Hence, in this study, Treg behaviour during IL-2-producing immune responses was explored using in vivo and in vitro murine systems. The use of murine mixed lymphocyte culture (MLC) revealed that a large proportion of Tregs increased in size, accompanied by both cell death and proliferation status. Further, these large Tregs, which were found to not recognize specific antigens, were observed in MLCs as being functionally activated by various cytokines, including IL-2, produced by antigen-specific T cells. This 'bystander Treg activation' was also observed in mice with graft-versus-host reactions (GvHRs). Alternatively, effector cells from Treg-depleted MLCs exhibited lower antigen-specific responses or higher cross-reactivity as compared to control MLCs with Tregs. Taken together, these results suggest that Tregs are activated by cytokines, mainly IL-2, released from T cells that are activated by a specific antigen. Subsequently, these activated bystander Tregs contribute to the fine-tuning of highly antigen-specific immune responses.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Apoptosis , Biomarkers , Cell Death , Cell Size , Cytokines/metabolism , Immunophenotyping , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
13.
Immunol Lett ; 224: 21-27, 2020 08.
Article in English | MEDLINE | ID: mdl-32492473

ABSTRACT

Intestinal IgA is induced by microbes and food antigens. Peyer's patches (PPs) are known as one of the inductive sites for intestinal IgA production. However, the precise mechanism of IgA induction is as yet unknown. IgA secretion was induced from IgD+ B cells in vitro by stimulus with lipopolysaccharide in the presence of only retinoic acid (RA) and low doses of TGF-ß1. Surface IgA+ cells were effectively induced from IgD+ B cells in vitro by the mixture of RA and the cytokines TGF-ß1, APRIL, IL-5 and IL-21. rIL-21 upregulated surface IgA+ but impaired the proliferation of stimulated B cells in the presence of rTGF-ß1, RA and rAPRIL, in vitro. The addition of rIL-5 restored the impaired proliferation by rIL-21, resulting in the expansion of IgA+ cells. rIL-21 induced the expression of Aicda and Prdm1, and impaired Rel in IgD+ B cells. Blockade of IL-21R signaling by a neutralizing mAb in vivo led to lower frequencies of IgA+ and IgG2b+ cells and lower germinal center B cells in PPs in a homeostatic condition. Although amounts of small intestinal IgA and titers of anti-dsDNA, the major target of intestinal IgA, in these mice were not altered, anti-OVA IgA titers induced by OVA drinking in OVA-specific T-cell receptor (TCR) transgenic mice were decreased. PP-deficient TCR transgenic mice showed diminished anti-OVA IgA induction. Blockade of IL-5R signaling in vivo led to similar results with relatively weaker effects than that of IL-21R mAb administration. These results suggest that IL-21 and IL-5 play cooperative roles in surface expression of IgA in PPs.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulin A/metabolism , Interleukin-5/metabolism , Interleukins/metabolism , Peyer's Patches/immunology , Animals , Cells, Cultured , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Gene Expression Regulation , Immunoglobulin D/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Tretinoin/metabolism
14.
Immunol Lett ; 211: 53-59, 2019 07.
Article in English | MEDLINE | ID: mdl-31145915

ABSTRACT

Peyer's patches (PPs) are inductive sites for IgA production; however, the induction mechanism of IgA remains largely unknown. We found that the activated phenotypes of isotype-switched PP B cells were more abundant than those of splenic B cells. Immunoglobulins (Igs) from PP B cells reacted to several substances, including DNA and diet extract. Hybridomas established from PP B cells of untreated mice revealed that IgA mainly react with DNA. PP-deficient mice revealed that PPs were dispensable for a total intestinal IgA amount but were required for intestinal anti-diet extract and anti-DNA IgA. Antibiotic-treated mice and CD4+ T cell-depleted mice demonstrated that the intestinal anti-DNA IgA was induced by microbiota in a T-independent manner. Interestingly, the oral administration of IgA led to the expansion of intestinal bacteria in a reactivity-independent manner. Our findings suggest that PPs are unique and efficient inductive sites for IgA, particularly against T-independent antigens.


Subject(s)
Antibodies, Antinuclear/metabolism , B-Lymphocytes/immunology , Gastrointestinal Microbiome/immunology , Immunoglobulin A/metabolism , Intestines/immunology , Peyer's Patches/immunology , Animals , Cells, Cultured , Female , Hybridomas , Immunoglobulin Class Switching , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes/immunology
15.
Cell Immunol ; 323: 41-48, 2018 01.
Article in English | MEDLINE | ID: mdl-29100594

ABSTRACT

Intestinal immunoglobulins (Igs) protect against microbes. However, the regulation of intestinal Ig production is poorly understood. In this study, we have investigated the roles of APRIL (tumor necrosis factor superfamily member [TNFSF] 13) and BAFF (TNFSF13B) in intestinal Ig induction. Peyer's patches (PPs) are, at least in part, an inductive site for Igs, including IgA. Introducing APRIL and BAFF in vivo lowered the frequency of IgG1+ or IgG2b+ B cells in PPs. Administration of TACI-Fc upregulated the frequency of IgG1+, IgG2b+, and IgA+ B cells in PPs, suggesting that APRIL and BAFF attenuate Ig production in these regions. TACI-Fc also upregulated intestinal IgA levels and expanded germinal center B cells in PPs. These results indicate that APRIL and BAFF paradoxically downregulate homeostatic Ig production in the intestines.


Subject(s)
B-Cell Activating Factor/metabolism , Immunoglobulins/biosynthesis , Intestines/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , Antibody Formation , B-Cell Activating Factor/immunology , B-Cell Activation Factor Receptor/metabolism , B-Cell Maturation Antigen/metabolism , B-Lymphocytes/immunology , Down-Regulation , Female , Homeostasis/immunology , Immunoglobulin A/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulins/metabolism , Male , Mice , Mice, Inbred C57BL , Peyer's Patches/immunology , Tumor Necrosis Factor Ligand Superfamily Member 13/immunology , Tumor Necrosis Factor-alpha/metabolism
16.
Immunol Lett ; 165(1): 1-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25794634

ABSTRACT

Anatomical containment of commensal bacteria in the intestinal mucosa is promoted by innate lymphoid cells (ILCs). However, the mechanism by which ILCs regulate bacterial localization to specific regions remains unknown. Here we show that Peyer's patch (PP) ILCs robustly produce IL-22 and IFN-γ in the absence of exogenous stimuli. Antibiotic treatment of mice decreased both IL-22+ and IFN-γ+ cells in PPs. Blockade of both IL-2 and IL-23 signaling in vitro lowered IL-22 and IFN-γ production. PP ILCs induced mRNA expression of the antibacterial proteins RegIIIß and RegIIIγ in intestinal epithelial cells. Furthermore, in vivo depletion of ILCs rather than T cells altered bacterial composition and allowed bacterial proliferation in PPs. Collectively, our results show that ILCs regulate the expansion of commensal bacteria in PPs.


Subject(s)
Bacterial Physiological Phenomena/immunology , Immunity, Innate/physiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocyte Subsets/immunology , Peyer's Patches/cytology , Peyer's Patches/immunology , Animals , Antigens, Surface/metabolism , Bacteria , Bacterial Physiological Phenomena/drug effects , Cytokines/biosynthesis , Gastrointestinal Microbiome/immunology , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , Intestinal Mucosa/metabolism , Lymphocyte Subsets/metabolism , Mice , Pancreatitis-Associated Proteins , Proteins/metabolism , Signal Transduction , Interleukin-22
17.
Eur J Immunol ; 45(3): 876-85, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25504587

ABSTRACT

Eosinophils are multifunctional leukocytes involved in allergic reactions as well as adipose tissue regulation. IL-5 is required for eosinophil survival; however, the in vivo mechanisms of eosinophil regulation are not fully understood. A tg mouse model with il5 promoter-driven EGFP expression was established for detecting the IL-5-producing cells in vivo. Il5-egfp tg mice expressed high levels of EGFP in gonadal adipose tissue (GAT) cells. EGFP(+) cells in GAT were mainly group 2 innate lymphoid cells (ILCs). IL-33 preferentially expanded EGFP(+) cells and eosinophils in GAT in vivo. EGFP(+) ILCs were found to upregulate prg2 mRNA expression in GAT eosinophils. These results demonstrate that ILCs activate eosinophils in GAT. The blockage of IL-33Rα, on the other hand, did not impair EGFP(+) ILC numbers but did impair eosinophil numbers in vivo. GAT eosinophils expressed IL-33Rα and IL-33 expanded eosinophil numbers in CD90(+) cell-depleted mice. IL-33 was further observed to induce the expression of retnla and epx mRNA in eosinophils. These findings demonstrate that IL-33 directly activates eosinophils in GAT, and together with our other findings described above, our findings show that IL-33 has dual pathways via which it activates eosinophils in vivo: a direct activation pathway and a group 2 ILC-mediated pathway.


Subject(s)
Eosinophils/immunology , Gene Expression Regulation/immunology , Immunity, Innate/physiology , Interleukins/immunology , Intra-Abdominal Fat/immunology , Animals , Eosinophil Major Basic Protein/genetics , Eosinophil Major Basic Protein/immunology , Eosinophils/cytology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-33 , Interleukin-5/genetics , Interleukin-5/immunology , Interleukins/genetics , Intra-Abdominal Fat/cytology , Mice , Mice, Knockout
18.
Cytotechnology ; 66(3): 373-82, 2014 May.
Article in English | MEDLINE | ID: mdl-23934330

ABSTRACT

IL-5 plays important roles in eosinophil differentiation, expansion, and recruitment. The regulation of IL-5 seems critical for the treatment of eosinophil-mediated allergic reactions. However, the precise mechanisms for IL-5 regulation remain unknown. In this study, we investigated how IL-5 production is regulated. The transduction of GATA-3 into a murine T cell hybridoma resulted in acquiring the ability to produce IL-5 in response to an antigenic stimulus like Th2 cells. This production was dependent on the cAMP-PKA pathway, but not on p38 activation. Transduction of NIK largely impaired IL-5 production. RelA and RelB similarly impaired IL-5 production. RelA decreased not only IL-5 protein amount but mRNA. RelA also inhibited Il5-luciferase reporter activity. The transduction of GATA-3 decreased the expression of Tbx21 and Eomes, but the additional transduction of RelA abrogated the decreased expression of GATA-3-induced Tbx21 and Eomes. Furthermore, the transduction of T-bet or Eomes into the GATA-3-transduced T cell hybridoma impaired IL-5 production. These results suggested that strong enhancement of the NFκB pathway downregulates IL-5 production and upregulates T-box protein expression to shift an immune response from Th2 to inflammatory Th1.

19.
Thromb Res ; 131(5): 444-9, 2013 May.
Article in English | MEDLINE | ID: mdl-23566532

ABSTRACT

INTRODUCTION: Factor VIII (FVIII) treatment for hemophilia A has difficulties in correcting bleeding diathesis in the presence of inhibitors. MATERIALS AND METHODS: An adeno-associated virus type 8 (AAV8) vector containing the factor VII (FVII) gene or the activated factor VII (FVIIa) gene was used to investigate the therapeutic effect of FVII or FVIIa overexpression in FVIII-deficient mice with inhibitors. RESULTS: Following repeated human FVIII injection, FVIII-deficient mice developed anti-human FVIII antibodies that cross-reacted with mouse FVIII. High transgene expression of murine FVII or murine FVIIa was achieved using the AAV8 vector and resulted in increased blood FVII activity greater than 800% of normal murine FVII levels in vector-injected FVIII-deficient mice. Thromboelastography analysis showed significant improvements in clotting time, clot formation time, α angle, and mean clot firmness in AAV8 vector-injected FVIII-deficient mice with inhibitors. Overexpression of FVIIa ameliorated the bleeding phenotype of FVIII-deficient mice with inhibitors and significantly increased the survival rate after tail clipping. In addition, overexpression of FVII increased the survival rate of FVIII-deficient mice with inhibitors after tail clipping though it was not as efficient as FVIIa overexpression. CONCLUSIONS: These data suggest that FVII overexpression is an alternative strategy for the treatment of hemophilia A with inhibitors.


Subject(s)
Factor VII/biosynthesis , Hemorrhage/therapy , Animals , Dependovirus/genetics , Factor VII/genetics , Factor VIIa/biosynthesis , Factor VIIa/genetics , Genetic Therapy , Hemophilia A/drug therapy , Hemophilia A/genetics , Hemophilia A/immunology , Hemophilia A/therapy , Hemorrhage/drug therapy , Hemorrhage/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Survival Rate , Transfection
20.
Hum Gene Ther ; 24(3): 283-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23360488

ABSTRACT

Hemophilia is an X-linked bleeding disorder, and patients with hemophilia are deficient in a biologically active coagulation factor. This study was designed to combine the efficiency of lentiviral vector transduction techniques with murine adipose tissue-derived stem/stromal cells (mADSCs) as a new method to produce secreted human coagulation factor IX (hFIX) and to treat hemophilia B. mADSCs were transduced with simian immunodeficiency virus (SIV)-hFIX lentiviral vector at multiplicities of infection (MOIs) from 1 to 60, and the most effective dose was at an MOI of 10, as determined by hFIX production. hFIX protein secretion persisted over the 28-day experimental period. Cell sheets composed of lentiviral vector-transduced mADSCs were engineered to further enhance the usefulness of these cells for future therapeutic applications in transplantation modalities. These experiments demonstrated that genetically transduced ADSCs may become a valuable cell source for establishing cell-based gene therapies for plasma protein deficiencies, such as hemophilia.


Subject(s)
Adipose Tissue/cytology , Genetic Therapy , Genetic Vectors/genetics , Hemophilia B/genetics , Hemophilia B/therapy , Mesenchymal Stem Cells/metabolism , Simian Immunodeficiency Virus/genetics , Animals , Blood Coagulation , Factor IX/biosynthesis , Factor IX/genetics , Gene Order , Hemophilia B/blood , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Mice , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...