Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Ke-Qin Xin is active.

Publication


Featured researches published by Ke-Qin Xin.


Proceedings of the National Academy of Sciences of the United States of America | 2007

The Atg5 Atg12 conjugate associates with innate antiviral immune responses.

Nao Jounai; Fumihiko Takeshita; Kouji Kobiyama; Asako Sawano; Atsushi Miyawaki; Ke-Qin Xin; Ken J. Ishii; Taro Kawai; Shizuo Akira; Koichi Suzuki; Kenji Okuda

Autophagy is an essential process for physiological homeostasis, but its role in viral infection is only beginning to be elucidated. We show here that the Atg5–Atg12 conjugate, a key regulator of the autophagic process, plays an important role in innate antiviral immune responses. Atg5-deficient mouse embryonic fibroblasts (MEFs) were resistant to vesicular stomatitis virus replication, which was largely due to hyperproduction of type I interferons in response to immunostimulatory RNA (isRNA), such as virus-derived, double-stranded, or 5′-phosphorylated RNA. Similar hyperresponse to isRNA was also observed in Atg7-deficient MEFs, in which Atg5–Atg12 conjugation is impaired. Overexpression of Atg5 or Atg12 resulted in Atg5–Atg12 conjugate formation and suppression of isRNA-mediated signaling. Molecular interaction studies indicated that the Atg5–Atg12 conjugate negatively regulates the type I IFN production pathway by direct association with the retinoic acid-inducible gene I (RIG-I) and IFN-β promoter stimulator 1 (IPS-1) through the caspase recruitment domains (CARDs). Thus, in contrast to its role in promoting the bactericidal process, a component of the autophagic machinery appears to block innate antiviral immune responses, thereby contributing to RNA virus replication in host cells.


Journal of Immunology | 2000

The Timing of GM-CSF Expression Plasmid Administration Influences the Th1/Th2 Response Induced by an HIV-1-Specific DNA Vaccine

Ken-ichi Kusakabe; Ke-Qin Xin; Hidenori Katoh; Kaharu Sumino; Eri Hagiwara; Susumu Kawamoto; Katsuji Okuda; Yohei Miyagi; Ichiro Aoki; Kusuya Nishioka; Dennis M. Klinman; Kenji Okuda

The mechanism of immune activation induced by a plasmid-encoding GM-CSF (pGM-CSF), administered in combination with a DNA vaccine encoding the envelope of HIV, was studied. Injecting pGM-CSF i.m. into mice 3 days before DNA vaccination primarily induced a Th2 response. Simultaneous administration of the DNA vaccine plus pGM-CSF activated both a Th1 and a Th2 response. When the plasmid was injected 3 days after DNA vaccination, enhancement of Th1 immunity predominated. These results suggest that the timing of cytokine expression determines the phenotype of the resultant Th response. After 3 days of pGM-CSF injection, the increased percentages of CD11c+, CD8+ cells were observed in the regional lymph nodes. In addition, many infiltrated cells, including S-100 protein-positive cells, were found in the pGM-CSF-injected tissue. The importance of these S-100+ cells or both CD8+ and CD11c+ cells, especially that of dendritic cells (DCs), was also studied. DCs derived from bone marrow and cultured in RPMI 1640 medium containing IL-4 and GM-CSF were incubated with DNA vaccine and then transferred into naive mice. Mice receiving DCs showed strong HIV-1-specific Th2 immune responses. Our results suggest that DCs play important roles in the activation or modification of the Th2-type immune response induced by DNA vaccination.


Vaccine | 2001

Protective immunity against influenza A virus induced by immunization with DNA plasmid containing influenza M gene.

Kenji Okuda; Atsushi Ihata; Setsuko Watabe; Eiichi Okada; Tadashi Yamakawa; Kenji Hamajima; Jun Yang; Norihisa Ishii; Masatoshi Nakazawa; Katsuji Okuda; Katsuhiro Ohnari; Katsuhisa Nakajima; Ke-Qin Xin

DNA vaccination is characterized by its preferential induction of the cytotoxic T cell lymphocyte (CTL) response and is expected to be a useful means of protection against viral infection. We examined the protective effect of an expression plasmid (pME18S-M) containing M1 and M2 genes of influenza A/PR/8/34. We detected the CTL activity by introducing these plasmids into BALB/c mice by either the intramuscular or the intranasal route. The influenza-specific antibody response was also induced, although its neutralizing effect against influenza virus was not observed. From 70 to 80% protection was observed in the mice immunized with the pME18S-M plasmid followed by lethal infection with influenza viruses of the A/WSN/33 and A/PR/8/34 strains, whereas all mice without the plasmid vaccination failed to survive. This protective activity was significantly weakened when the CD8(+) cells of these immunized mice were eliminated by several injections of anti-CD8 antibody. The protective activity was also weakened when anti-CD4 antibody was injected in the early phase of DNA vaccination. These data suggest that the pME18S-M plasmid is useful as a DNA vaccine for overcoming highly mutational influenza viruses.


Vaccine | 2002

Adjuvant effect of multi-CpG motifs on an HIV-1 DNA vaccine

Yoshitsugu Kojima; Ke-Qin Xin; Takaaki Ooki; Kenji Hamajima; Tomohiro Oikawa; Kaori Shinoda; Tomomi Ozaki; Yuka Hoshino; Nao Jounai; Masatoshi Nakazawa; Dennis M. Klinman; Kenji Okuda

Synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs trigger an immune response characterized by the activation of B cells, NK cells and monocytes/macrophages. Based on evidence that the immunogenicity of DNA vaccines can be augmented by the addition of CpG motifs, 5-20 additional CpG motifs were cloned into a pUC-derived plasmid. Treating bone-marrow derived dendritic cells (BM-DCs) with CpG-enriched plasmids in vitro boosted their expressions of MHC class II molecules, the CD40 and CD86 activation markers. Co-administering the CpG-enriched plasmids with a DNA vaccine encoding the envelope glycoprotein of HIV to BALB/c mice significantly increased HIV-specific cell mediated and humoral immunity. A significant boost was observed when the CpG plasmid was administered either 2 or 4 days after DNA vaccination. Plasmids containing 20 CpG copies were the most effective immune enhancers both in vitro and in vivo. These results suggest that plasmids containing multiple CpG motifs may improve the immunogenicity of DNA vaccines.


Vaccine | 1999

IL-15 expression plasmid enhances cell-mediated immunity induced by an HIV-1 DNA vaccine

Ke-Qin Xin; Kenji Hamajima; Shin Sasaki; Takashi Tsuji; Setsuko Watabe; Eiichi Okada; Kenji Okuda

Cytokines are powerful regulators of the immune response. In this study, an HIV-1 envelope DNA vaccine and interleukin 15 (IL-15) expression plasmid were intranasally administered to mice. A significant increase in the HIV-1-specific DTH response and CTL activity, and decrease in the serum IgG/IgG2a ratio was observed in the group which received DNA vaccine and IL-15 expression plasmid compared to DNA vaccination alone. Restimulated immune lymphoid cells from mice which received both agents showed enhanced production of interferon-gamma (IFN-gamma) and reduced secretion of IL-4. However, administration of DNA vaccine with IL-15 and IL-2 or IL-12 expression plasmids did not alter the effect of IL-15 expression plasmid on the DNA vaccine. These results indicate that intranasal administration of DNA vaccine and IL-15 expression plasmid is capable of enhancing the T helper type 1 (Th1) dependent HIV-1-specific cell-mediated immunity, and that the IL-15 and IL-2 or IL-12 expression plasmids may not have a synergistic effect on the immune response induced by DNA vaccine in vivo.


Human Gene Therapy | 2001

A novel recombinant adeno-associated virus vaccine induces a long-term humoral immune response to human immunodeficiency virus

Ke-Qin Xin; Masashi Urabe; Jun Yang; Kazuo Nomiyama; Hiroaki Mizukami; Kenji Hamajima; Hiroko Nomiyama; Tomoyuki Saito; Mitsunobu Imai; John Monahan; Katsuji Okuda; Keiya Ozawa; Kenji Okuda

Recombinant adeno-associated virus (AAV) has attracted tremendous interest as a promising vector for gene delivery. In this study we have developed an HIV-1 vaccine, using an AAV vector expressing HIV-1 env, tat, and rev genes (AAV-HIV vector). A single injection of the AAV-HIV vector induced strong production of HIV-1-specific serum IgG and fecal secretory IgA antibodies as well as MHC class I-restricted CTL activity in BALB/c mice. The titer of HIV-1-specific serum IgG remained stable for 10 months. When AAV-HIV vector was coadministered with AAV-IL2 vector, the HIV-specific cell-mediated immunity (CMI) was significantly enhanced. Boosting with AAV-HIV vector strongly enhanced the humoral response. Furthermore, the mouse antisera neutralized an HIV-1 homologous strain, and BALB/c mice immunized via the intranasal route with an AAV vector expressing the influenza virus hemagglutinin (HA) gene showed protective immunity against homologous influenza virus challenge. These results demonstrate that AAV-HIV vector immunization may provide a novel and promising HIV vaccination strategy.


Methods | 2003

Adjuvant formulations and delivery systems for DNA vaccines.

Shin Sasaki; Fumihiko Takeshita; Ke-Qin Xin; Norihisa Ishii; Kenji Okuda

DNA vaccines have become a reliable and major means to elicit immune responses in the past decade. We and others have attempted to obtain stronger, more long lasting, and optimized immune responses, subsequent to the pioneering works demonstrating the ability of plasmid DNA to raise specific immune responses. Advances in molecular biology and biotechnology allow the application of various adjuvants, immunologic agents that increase the antigenic response, in DNA vaccines. Adjuvants can be broadly separated into two classes based on their origin-genetic and conventional. Genetic adjuvants are expression vectors of cytokines or other molecules that can modulate immune responses when administered with a vaccine antigen. Conventional adjuvants are chemical compounds that enhance, prolong, or modulate antigen-specific immune responses. The use of an appropriate adjuvant is pivotal in optimizing the response to DNA vaccines. Moreover, DNA vaccines themselves possess their own adjuvant activity because of the presence of unmethylated CpG motifs in particular base contents. The route of inoculation is also a critical factor in determining the outcome of vaccination. It is well known that intramuscular injection preferentially induces Th1-type immunity, whereas particle bombardment by gene gun predominantly induces Th2-type response. This article focuses on providing the detailed procedure to construct genetic adjuvant plasmids and prepare DNA vaccines formulated with conventional adjuvants. We also offer a practical guide for the procedure of intramuscular DNA injection.


Vaccine | 2001

Protection against influenza virus challenge by topical application of influenza DNA vaccine.

Setsuko Watabe; Ke-Qin Xin; Atsushi Ihata; Li-Juan Liu; Akiko Honsho; Ichiro Aoki; Kenji Hamajima; Britta Wahren; Kenji Okuda

We studied the use of a DNA vaccine expressing the matrix (M) gene of the influenza virus A/PR/8/34. Mice were immunized by painting the DNA vaccine three times on the skin after removal of its keratinocytic layers. Immunization by this method produced M-specific antibodies and cytotoxic T lymphocyte (CTL) response, and acquired resistance against influenza virus challenge. This protection was abrogated by the in vivo injection of anti-CD8 or anti-CD4 monoclonal antibody. We further found that simultaneous topical application (t.a.) of GM-CSF expression plasmid (pGM-CSF) or liposomes plus mannan produced stronger immune response competence and enhanced the protective effect against influenza virus challenge. The present study revealed that administering DNA vaccine by topical application can elicit both humoral and cell-mediated immunity (CMI).


Journal of Virology | 2006

Scalable Generation of High-Titer Recombinant Adeno-Associated Virus Type 5 in Insect Cells

Masashi Urabe; Takayo Nakakura; Ke-Qin Xin; Yoko Obara; Hiroaki Mizukami; Akihiro Kume; Robert M. Kotin; Keiya Ozawa

ABSTRACT We established a method for production of recombinant adeno-associated virus type 5 (rAAV5) in insect cells by use of baculovirus expression vectors. One baculovirus harbors a transgene between the inverted terminal repeat sequences of type 5, and the second expresses Rep78 and Rep52. Interestingly, the replacement of type 5 Rep52 with type 1 Rep52 generated four times more rAAV5 particles. We replaced the N-terminal portion of type 5 VP1 with the equivalent portion of type 2 to generate infectious AAV5 particles. The rAAV5 with the modified VP1 required α2-3 sialic acid for transduction, as revealed by a competition experiment with an analog of α2-3 sialic acid. rAAV5-GFP/Neo with a 4.4-kb vector genome produced in HEK293 cells or Sf9 cells transduced COS cells with similar efficiencies. Surprisingly, Sf9-produced humanized Renilla green fluorescent protein (hGFP) vector with a 2.4-kb vector genome induced stronger GFP expression than the 293-produced one. Transduction of murine skeletal muscles with Sf9-generated rAAV5 with a 3.4-kb vector genome carrying a human secreted alkaline phosphatase (SEAP) expression cassette induced levels of SEAP more than 30 times higher than those for 293-produced vector 1 week after injection. Analysis of virion DNA revealed that in addition to a 2.4- or 3.4-kb single-stranded vector genome, Sf9-rAAV5 had more-abundant forms of approximately 4.7 kb, which appeared to correspond to the monomer duplex form of hGFP vector or truncated monomer duplex SEAP vector DNA. These results indicated that rAAV5 can be generated in insect cells, although the difference in incorporated virion DNA may induce different expression patterns of the transgene.


Clinical and Experimental Immunology | 1999

Macrophage inflammatory protein-1α (MIP-1α) expression plasmid enhances DNA vaccine-induced immune response against HIV-1

Lu Y; Ke-Qin Xin; Kenji Hamajima; Takashi Tsuji; Ichiro Aoki; Jun Yang; Shin Sasaki; Jun Fukushima; T. Yoshimura; S. Toda; Eiichi Okada; Kenji Okuda

CD8+ cell‐secreted CC‐chemokines, MIP‐1α, and MIP‐β have recently been identified as factors which suppress HIV. In this study we co‐inoculated MIP‐1α expression plasmid with a DNA vaccine constructed from HIV‐1 pCMV160IIIB and pcREV, and evaluated the effect of the adjuvant on HIV‐specific immune responses following intramuscular and intranasal immunization. The levels of both cytotoxic T lymphocyte (CTL) activity and DTH showed that HIV‐specific cell‐mediated immunity (CMI) was significantly enhanced by co‐inoculation of the MIP‐1α expression plasmid with the DNA vaccine compared with inoculation of the DNA vaccine alone. The HIV‐specific serum IgG1/IgG2a ratio was significantly lowered when the plasmid was co‐inoculated in both intramuscular and intranasal routes, suggesting a strong elicitation of the T helper (Th) 1‐type response. When the MIP‐1α expression plasmid was inoculated intramuscularly with the DNA vaccine, an infiltration of mononuclear cells was observed at the injection site. After intranasal administration, the level of mucosal secretory IgA antibody was markedly enhanced. These findings demonstrate that MIP‐1α expression plasmid inoculated together with DNA vaccine acts as a strong adjuvant for eliciting Th1‐derived immunity.

Collaboration


Dive into the Ke-Qin Xin's collaboration.

Top Co-Authors

Avatar

Kenji Okuda

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar

Kenji Hamajima

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar

Dennis M. Klinman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nao Jounai

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar

Jun Fukushima

Akita Prefectural University

View shared research outputs
Top Co-Authors

Avatar

Shin Sasaki

Yokohama City University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge