Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 13033, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563266

ABSTRACT

Emerging SARS-CoV-2 Omicron variants are highly contagious with enhanced immune escape mechanisms against the initially approved COVID-19 vaccines. Therefore, we require stable alternative-platform vaccines that confer protection against newer variants of SARS-CoV-2. We designed an Omicron B.1.1.529 specific DNA vaccine using our DNA vaccine platform and evaluated the humoral and cellular immune responses. SD rats intradermally administered with Omicron-specific DNA vaccine via pyro-drive jet injector (PJI) thrice at 2-week intervals elicited high antibody titers against the Omicron subvariants as well as the ancestral strain. Indeed, the Omicron B.1.1.529-specific antibody titer and neutralizing antibody were higher than that of other strains. Longitudinal monitoring indicated that anti-spike (ancestral and Omicron) antibody titers decreased toward 30 weeks after the first vaccination dose. However, neutralization activity remained unaltered. Germinal center formation was histologically detected in lymph nodes in rats immunized with Omicron DNA vaccine. Ancestral spike-specific immune cell response was slightly weaker than Omicron spike-specific response in splenocytes with Omicron-adapted DNA vaccine, evaluated by ELISpot assay. Collectively, our findings suggest that Omicron targeting DNA vaccines via PJI can elicit robust durable antibody production mediated by germinal center reaction against this new variant as well as partially against the spike protein of other SARS-CoV-2 variants.


Subject(s)
COVID-19 , Vaccines, DNA , Animals , Humans , Rats , Rats, Sprague-Dawley , Antibodies, Neutralizing , COVID-19 Vaccines , SARS-CoV-2 , COVID-19/prevention & control , Germinal Center , Antibodies, Viral
2.
Neurosci Lett ; 810: 137370, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37399972

ABSTRACT

Recent advances in endovascular treatment (EVT) dramatically changed the outcome of ischemic stroke, but partial reperfusion does not improve the outcome as well as no reperfusion. Although partial reperfusion is estimated to be more potential for therapeutic intervention than permanent occlusion due to some blood supply, their pathophysiological difference is still unknown. To answer the question, we analyzed the difference in mice, which were exposed to distal middle cerebral artery occlusion with 14-min common carotid artery (CCA) occlusion (partial reperfusion) or permanent CCA occlusion (no reperfusion). Although the final infarct volume was same between permanent and partial reperfusion, Fluoro-jade C staining showed that neurodegeneration was inhibited both in the severe and moderate ischemic region 3 h after partial reperfusion. Also, partial reperfusion increased the number of TUNEL-positive cells only in the severe ischemic region. IgG extravasation was suppressed at 24 h only in the moderate ischemic region in partial reperfusion. Injected FITC-dextran was observed in the brain parenchyma with BBB leakage at 24 h in partial reperfusion, but not in permanent occlusion. The expression of il1ß and il6 mRNA was inhibited in the severe ischemic region. Thus, partial reperfusion showed region-dependent favorable pathophysiology, such as delayed neurodegeneration, suppressed BBB destruction and inflammation, and potential for drug delivery, when compared to permanent occlusion. Further studies on the molecular differences and effectiveness of drugs will shed light on the development of novel treatments for partial reperfusion in ischemic stroke.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Mice , Animals , Ischemic Stroke/metabolism , Brain/metabolism , Brain Ischemia/metabolism , Reperfusion , Infarction, Middle Cerebral Artery/metabolism , Cerebrovascular Circulation , Stroke/metabolism
3.
FEBS Open Bio ; 13(3): 478-489, 2023 03.
Article in English | MEDLINE | ID: mdl-36651034

ABSTRACT

Cell-cell fusion involves the fusion of somatic cells into a single hybrid cell. It is not only a physiological process but also an important cell engineering technology which can be applied to various fields, such as regenerative medicine, antibody engineering, genetic engineering, and cancer therapy. There are three major methods of cell fusion: electrical cell fusion, polyethylene glycol (PEG) cell fusion, and virus-mediated cell fusion. Although PEG cell fusion is the most economical approach and does not require expensive instrumentation, it has a poor fusion rate and induces a high rate of cell cytotoxicity. To improve the fusion rate of the PEG method, we combined it with the pyro-drive jet injector (PJI). PJI provides instant pressure instead of cell agitation to increase the probability of cell-to-cell contact and shorten the distance between cells in the process of cell fusion. Here, we report that this improved fusion method not only decreased cell cytotoxicity during the fusion process, but also increased fusion rate compared with the conventional PEG method. Furthermore, we tested the functionality of cells fused using the PJI-PEG method and found them to be comparable to those fused using the conventional PEG method in terms of their application for dendritic cell (DC)-tumor cell fusion vaccine production; in addition, the PJI-PEG method demonstrated excellent performance in hybridoma cell preparation. Taken together, our data indicate that this method improves cell fusion efficiency as compared to the PEG method and thus has the potential for use in various applications that require cell fusion technology.


Subject(s)
Genetic Engineering , Polyethylene Glycols , Polyethylene Glycols/pharmacology , Cell Fusion
4.
Sci Rep ; 12(1): 20923, 2022 12 03.
Article in English | MEDLINE | ID: mdl-36463322

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has led to a global pandemic. New technologies have been utilized to develop several types of vaccines to prevent the spread of SARS-CoV-2 infection, including mRNA vaccines. Our group previously developed an effective DNA-based vaccine. However, emerging SARS-CoV-2 variants of concern (VOCs), such as the delta variant, have escaped mutations against vaccine-induced neutralizing antibodies. This suggests that modified vaccines accommodating VOCs need to be developed promptly. Here, we first modified the current DNA vaccine to enhance antigenicity. Compared with the parental DNA vaccine, the modified version (GP∆-DNA vaccine) induced rapid antibody production. Next, we updated the GP∆-DNA vaccine to spike glycoprotein of the delta variant (GP∆-delta DNA vaccine) and compared the efficacy of different injection routes, namely intramuscular injection using a needle and syringe and intradermal injection using a pyro-drive jet injector (PJI). We found that the levels of neutralizing antibodies induced by the intradermal PJI injection were higher than intramuscular injection. Furthermore, the PJI-injected GP∆-delta DNA vaccine effectively protected human angiotensin-converting enzyme 2 (hACE2) knock-in mice from delta-variant infection. These results indicate that the improved DNA vaccine was effective against emerging VOCs and was a potential DNA vaccine platform for future VOCs or global pandemics.


Subject(s)
COVID-19 , Vaccines, DNA , Humans , Animals , Mice , SARS-CoV-2/genetics , Immunity, Humoral , Vaccines, DNA/genetics , COVID-19/prevention & control , Antibodies, Neutralizing
5.
Immunol Med ; 45(4): 251-264, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36001011

ABSTRACT

There is an urgent need to stop the coronavirus disease 2019 (COVID-19) pandemic through the development of efficient and safe vaccination methods. Over the short term, plasmid DNA vaccines can be developed as they are molecularly stable, thus facilitating easy transport and storage. pVAX1-SARS-CoV2-co was designed for the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) S protein. The antibodies produced led to immunoreactions against the S protein, an anti-receptor-binding-domain, and a neutralizing action of the pVAX1-SARS-CoV2-co, as previously confirmed. To promote the efficacy of the pVAX1-SARS-CoV2-co vaccine a pyro-drive jet injector (PJI) was used. An intradermally adjusted PJI demonstrated that the pVAX1-SARS-CoV2-co vaccine injection caused a high production of anti-S protein antibodies, triggered immunoreactions, and neutralized the actions against SARS-CoV-2. A high-dose pVAX1-SARS-CoV2-co intradermal injection using PJI did not cause any serious disorders in the rat model. A viral challenge confirmed that intradermally immunized mice were potently protected from COVID-19. A pVAX1-SARS-CoV2-co intradermal injection using PJI is a safe and promising vaccination method for overcoming the COVID-19 pandemic.


Subject(s)
COVID-19 , Vaccines, DNA , Viral Vaccines , Mice , Humans , Rats , Animals , COVID-19/prevention & control , Pandemics/prevention & control , SARS-CoV-2 , RNA, Viral , Rodentia , Antibodies, Viral , Vaccination/methods , Antibody Formation , Plasmids
6.
Mol Ther Oncolytics ; 20: 175-186, 2021 Mar 26.
Article in English | MEDLINE | ID: mdl-33575480

ABSTRACT

Breast cancer has a high risk of metastasis; however, no effective treatment has been established. We developed a novel immunotherapy for breast cancer to enhance cytotoxic T lymphocytes against cancer cells using N1-type neutrophils with anti-tumor properties. For this purpose, we combined CXCL2 (CXC chemokine ligand 2) plasmid DNA with inactivated Sendai virus (hemagglutinating virus of Japan)-envelope (HVJ-E). The combination of CXCL2 DNA and HVJ-E (C/H) suppressed the growth of murine breast cancers in orthotopic syngeneic models by enhancing cytotoxic T lymphocytes and inhibited lung metastasis of breast cancer from primary lesions. N1-type neutrophils (CD11b+ Ly6G+ FAS+) increased in the tumor microenvironment with C/H treatment, and tumor suppression and cytotoxic T lymphocyte activation from C/H was blocked after administrating anti-neutrophil antibodies, which indicates the role of N1-type neutrophils in cancer immunotherapy. We also demonstrated that the anti-tumor activities of C/H treatment were enhanced by the administration of anti-PD-1 antibodies through neutrophil-mediated cytotoxic T lymphocyte activation. Thus, the triple combination of C/H and anti-PD-1 antibody C/H treatment may provide an improvement in cancer immunotherapy.

7.
Cancer Gene Ther ; 27(6): 498-508, 2020 06.
Article in English | MEDLINE | ID: mdl-31383952

ABSTRACT

Inactivated Sendai virus particle (or hemagglutinating virus of Japan envelope; HVJ-E) has been previously reported to possess antitumour properties that activate antitumour immunity. Two glycoproteins, fusion (F) and hemagglutinin-neuraminidase (HN), are present on the surface of HVJ-E. HN is necessary for binding to receptors such as acidic gangliosides, and F induces membrane fusion by associating with membrane lipids. We previously reported that liposomes reconstituted with F but not HN showed antitumour activity by inducing IL-6 secretion in dendritic cells (DCs), suggesting that F protein is capable of eliciting antitumour activity. Here, we attempted to deliver F gene into tumour tissue in mice by electroporation and demonstrated that F gene therapy retarded tumour growth, increased CD4+ and CD8+ T-cell infiltration into tumours and induced tumour-specific IFN-γ T-cell response. However, neutralisation of IL-6R signalling did not impact F plasmid-mediated antitumour effect. Instead, we found that F plasmid treatment resulted in a significant increase in the secretion of the chemokine RANTES (regulated upon activation, normal T cell expressed and secreted) by tumour-infiltrating T cells. Neutralising antibody against RANTES abolished the antitumour effect of F plasmid treatment in a dose-dependent manner. Thus, F gene therapy may show promise as a novel therapeutic for single or combined cancer immunotherapy.


Subject(s)
Immunotherapy/methods , Melanoma, Experimental/therapy , Sendai virus/immunology , Animals , Female , Gene Fusion , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Plasmids/administration & dosage , Plasmids/genetics , Plasmids/immunology , Sendai virus/genetics , T-Lymphocytes/immunology , Transfection
8.
Oncotarget ; 7(27): 42195-42207, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27259252

ABSTRACT

The tumor microenvironment (TME) fosters tumors by attenuating anti-tumor immunity, reinforcing tumor cell survival and increasing angiogenesis. Among the constituents of the TME, here, we focused on tumor-associated neutrophils (TANs). First, we found that the combination of poly I:C and inactivated Sendai virus particles (hemagglutinating virus of Japan envelope; HVJ-E) synergistically suppressed tumor growth in the B16-F10 melanoma mouse model. In this model, poly I:C contributed to the recruitment of CD11b+Ly6G+ neutrophils to the TME, and co-injection of poly I:C and HVJ-E increased CD11b+Ly6G+FAS+ TAN in the TME. Depletion of neutrophils abolished the synergistic anti-tumor effect of HVJ-E and poly I:C in B16-F10 tumors. We revealed that C-X-C motif chemokine ligand 2 (CXCL2) is produced in the TME by poly I:C, but HVJ-E enhanced neutrophil infiltration of the TME does not occur. An anti-CXCL2 antibody inhibited the tumor suppression by HVJ-E+poly I:C. HVJ-E in combination with recombinant CXCL2 protein or CXCL2 pDNA suppressed mouse melanoma by increasing cytotoxic T lymphocyte activity against B16-F10 melanoma, which was abolished by an anti-Ly6G antibody. HVJ-E directly and indirectly increased FAS and ICAM-1 expression in cultured bone marrow-derived naïve neutrophils. Thus, HVJ-E activates anti-tumor immunity via anti-tumorigenic neutrophils in the TME. An HVJ-E vector containing the CXCL2 gene may be applicable as a novel cancer gene therapy strategy.


Subject(s)
Melanoma, Experimental/virology , Neutrophils/immunology , Neutrophils/virology , T-Lymphocytes/immunology , Tumor Microenvironment , Animals , Cell Line, Tumor , Cell Survival , Chemokine CXCL2/metabolism , Ligands , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Oncolytic Virotherapy , Poly I-C/immunology , Sendai virus , Signal Transduction , Spleen/cytology
9.
Vet Res ; 41(2): 15, 2010.
Article in English | MEDLINE | ID: mdl-19846041

ABSTRACT

Our previous report demonstrated that bovine ephemeral fever virus (BEFV)-infected cultured cells could induce caspase-dependent apoptosis. This study aims to further elucidate how BEFV activates the caspase cascade in bovine cells. BEFV replicated and induced apoptosis in Vero and Madin-Darby bovine kidney (MDBK) cells, and a kinetic study showed a higher efficiency of replication and a greater apoptosis induction ability of BEFV in Vero cells. Src and c-Jun N-terminal kinase (JNK) inhibitor, but not extracellular signal-regulated kinase (ERK) or p38 inhibitor, alleviated BEFV-mediated cytopathic effect and apoptosis. In BEFV-infected Vero and MDBK cells, BEFV directly induced Src tyrosine-418 phosphorylation and JNK phosphorylation and kinase activity, which was inhibited specifically by SU6656 and SP600125, respectively. The caspase cascade and its downstream effectors, Poly (ADP-ribose) polymerase (PARP) and DFF45, were also activated simultaneously upon BEFV infection. In addition, cytochrome c, but not Smac/DIABLO, was released gradually from mitochondria after BEFV infection. SU6656 suppressed Src, JNK, and caspase-3 and -9 activation, as well as PARP and DFF45 cleavage; SP600125 reduced JNK and caspase-3 and -9 activation, as well as PARP and DFF45 cleavage. Taken together, these results strongly support the hypothesis that a Src-dependent JNK signaling pathway plays a key role in BEFV-induced apoptosis. The molecular mechanism identified in our study may provide useful information for the treatment of BEFV.


Subject(s)
Apoptosis/physiology , Caspase 3/metabolism , Ephemeral Fever Virus, Bovine/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Kidney/cytology , Mitochondria/metabolism , Animals , Anthracenes/pharmacology , Caspase 3/genetics , Chlorocebus aethiops , Cytochromes c/metabolism , Dogs , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Indoles/pharmacology , Mitochondria/drug effects , Phosphorylation , Sulfonamides/pharmacology , Vero Cells , Virus Replication , src-Family Kinases/genetics , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...