Network


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

Hotspot


Dive into the research topics where Chi-Yuan Chen is active.

Publication


Featured researches published by Chi-Yuan Chen.


Biotechnology Advances | 2011

Baculovirus as a gene delivery vector: recent understandings of molecular alterations in transduced cells and latest applications.

Chi-Yuan Chen; Chin-Yu Lin; Guan-Yu Chen; Yu-Chen Hu

Abstract Baculovirus infects insects in nature and is non-pathogenic to humans, but can transduce a broad range of mammalian and avian cells. Thanks to the biosafety, large cloning capacity, low cytotoxicity and non-replication nature in the transduced cells as well as the ease of manipulation and production, baculovirus has gained explosive popularity as a gene delivery vector for a wide variety of applications. This article extensively reviews the recent understandings of the molecular mechanisms pertinent to baculovirus entry and cellular responses, and covers the latest advances in the vector improvements and applications, with special emphasis on antiviral therapy, cancer therapy, regenerative medicine and vaccine.


Biomaterials | 2010

The healing of critical-sized femoral segmental bone defects in rabbits using baculovirus-engineered mesenchymal stem cells

Chin-Yu Lin; Yu-Han Chang; Kun-Ju Lin; Tzu-Chen Yen; Ching-Lung Tai; Chi-Yuan Chen; Wen-Hsin Lo; Ing-Tsung Hsiao; Yu-Chen Hu

Management of massive segmental bone defects remains a challenging clinical problem and bone marrow-derived mesenchymal stem cells (BMSCs) hold promise for bone regeneration. To explore whether BMSCs engineered by baculovirus (an emerging gene delivery vector) can heal large bone defects, New Zealand White (NZW) rabbit BMSCs were transduced with the BMP2-expressing baculovirus or VEGF-expressing baculovirus, and co-implanted into critical-sized (10mm) femoral segmental defects in NZW rabbits. X-ray analysis revealed that the baculovirus-engineered BMSCs not only bridged the defects at as early as week 2, but also healed the defects in 100% of rabbits (13/13) at week 4. The osteogenic metabolism, as monitored by positron emission tomography (PET) also suggested the completion of bone healing at week 8. When compared with other control groups, the BMP2/VEGF-expressing BMSCs remarkably enhanced the segmental bone repair and mechanical properties, as evidenced by micro-computed tomography (microCT), histochemical staining and biomechanical testing. The ameliorated bone healing concurred with the augmented angiogenesis. These data demonstrated, that BMSCs engineered to express BMP2 and VEGF accelerate the repair of large femoral bone defects and improve the quality of the regenerated bone, which paves an avenue to utilizing baculovirus as a vector for BMSCs modification and regenerative medicine.


Journal of Virology | 2009

Baculovirus transduction of mesenchymal stem cells triggers the toll-like receptor 3 pathway.

Guan-Yu Chen; Hsiao-Chiao Shiah; Hung-Ju Su; Chi-Yuan Chen; Yung-Jen Chuang; Wen-Hsin Lo; Jie-Len Huang; Ching-Kuang Chuang; Shiaw-Min Hwang; Yu-Chen Hu

ABSTRACT Human mesenchymal stem cells (hMSCs) can be genetically modified with viral vectors and hold promise as a cell source for regenerative medicine, yet how hMSCs respond to viral vector transduction remains poorly understood, leaving the safety concerns unaddressed. Here, we explored the responses of hMSCs against an emerging DNA viral vector, baculovirus (BV), and discovered that BV transduction perturbed the transcription of 816 genes associated with five signaling pathways. Surprisingly, Toll-like receptor-3 (TLR3), a receptor that generally recognizes double-stranded RNA, was apparently upregulated by BV transduction, as confirmed by microarray, PCR array, flow cytometry, and confocal microscopy. Cytokine array data showed that BV transduction triggered robust secretion of interleukin-6 (IL-6) and IL-8 but not of other inflammatory cytokines and beta interferon (IFN-β). BV transduction activated the signaling molecules (e.g., Toll/interleukin-1 receptor domain-containing adaptor-inducing IFN-β, NF-κB, and IFN regulatory factor 3) downstream of TLR3, while silencing the TLR3 gene with small interfering RNA considerably abolished cytokine expression and promoted cell migration. These data demonstrate, for the first time, that a DNA viral vector can activate the TLR3 pathway in hMSCs and lead to a cytokine expression profile distinct from that in immune cells. These findings underscore the importance of evaluating whether the TLR3 signaling cascade plays roles in the immune response provoked by other DNA vectors (e.g., adenovirus). Nonetheless, BV transduction barely disturbed surface marker expression and induced only transient and mild cytokine responses, thereby easing the safety concerns of using BV for hMSCs engineering.


Vaccine | 2010

Enterovirus 71 virus-like particle vaccine: Improved production conditions for enhanced yield

Cheng-Yu Chung; Chi-Yuan Chen; Shih-Yeh Lin; Yao-Chi Chung; Hsin-Yi Chiu; Wei-Kuang Chi; Yu-Li Lin; Bor-Luen Chiang; Wei-Jheng Chen; Yu-Chen Hu

To develop the enterovirus 71 (EV71) vaccine, we previously constructed a recombinant baculovirus (Bac-P1-3CD) co-expressing EV71 P1 (under polyhedrin promoter) and 3CD (under p10 promoter) proteins, which caused P1 cleavage by 3CD protease and self-assembly of virus-like particles (VLPs) in Sf-9 cells. Assuming that reducing the 3CD expression can alleviate the competition with P1 expression and elevate the VLPs yield, hereby we constructed Bac-P1-C3CD and Bac-P1-I3CD expressing 3CD under weaker CMV and IE-1 promoters, respectively. Western blot and ELISA analyses revealed that Bac-P1-C3CD and Bac-P1-I3CD led to the VLPs release into the supernatant and enhanced the extracellular VLPs yield in Sf-9 cells, but gave poor VLPs production in High Five™ (Hi-5) cells. By optimizing the process parameters including host cells, cell density, culture mode and dissolved oxygen (DO), the best extracellular VLPs yield was achieved by infecting Sf-9 cells (4 × 10(6)cells/mL) cultured in the bioreactor (DO=30%) with Bac-P1-C3CD, which approached ≈64.3mg/L and represented a ≈43-fold increase over the yield (1.5mg/L) attained using the old process (Bac-P1-3CD infection of Sf-9 cells in the spinner flasks). The resultant VLPs not only resembled the VLPs produced from Bac-P1-3CD infection in density, size and shape, but also induced potent antibody responses in mouse models. The antibodies neutralized EV71 strains of homologous and heterologous genogroups, implicating the potential of the VLPs to confer cross-protection for the prevention of future epidemics. Altogether, Bac-P1-C3CD and the bioprocess render mass production more economical, obviate the need for cell lysis and hold promise for future industrial vaccine production.


Molecular Therapy | 2009

Development of a Hybrid Baculoviral Vector for Sustained Transgene Expression

Wen-Hsin Lo; Shiaw-Min Hwang; Ching-Kuang Chuang; Chi-Yuan Chen; Yu-Chen Hu

Baculovirus is a promising gene delivery vector but its widespread application is impeded as it only mediates transient transgene expression in mammalian cells. To prolong the expression, we developed a dual baculovirus system whereby one baculovirus expressed FLP recombinase while the other harbored an Frt-flanking cassette encompassing the transgene and oriP/EBNA1 derived from Epstein-Barr virus. After cotransduction of cells, the expressed FLP cleaved the Frt-flanking cassette off the baculovirus genome and catalyzed circular episome formation, then oriP/EBNA1 within the cassette enabled the self-replication of episomes. The excision/recombination efficiency was remarkably enhanced by sodium butyrate, reaching 75% in human embryonic kidney-293 (HEK293) cells, 85% in baby-hamster kidney (BHK) cells, 77% in primary chondrocytes, and 48% in mesenchymal stem cells (MSCs). The hybrid baculovirus substantially prolonged the transgene expression to approximately 48 days without selection and >63 days with selection, thanks to the maintenance of replicons and transgene transcription. In contrast to the replicating episomes, the baculovirus genome was rapidly degraded. Furthermore, an osteoinductive growth factor gene was efficiently delivered into MSCs using this system, which not only prolonged the growth factor expression but also potentiated the osteogenesis of MSCs. These data collectively implicate the potential of this hybrid baculovirus system in gene therapy applications necessitating sustained transgene expression.


Molecular Pharmaceutics | 2011

Biosafety Assessment of Human Mesenchymal Stem Cells Engineered by Hybrid Baculovirus Vectors

Chi-Yuan Chen; Hsiao-Hsuan Wu; Chih-Ping Chen; Schu-Rern Chern; Shiaw-Min Hwang; Shiu-Feng Huang; Wen-Hsin Lo; Guan-Yu Chen; Yu-Chen Hu

Mesenchymal stem cells (MSCs) hold promise for cell therapy, and implantation of MSCs engineered with a baculovirus transiently expressing the growth factor can augment the bone repair. To prolong the baculovirus-mediated transgene expression, we developed hybrid baculovirus vectors exploiting the FLP/Frt-mediated recombination for circular episome formation. Transduction of human MSCs with the hybrid baculovirus vectors harboring the osteoinductive bmp2 gene substantially extended the BMP2 expression and improved the cellular osteogenic differentiation. To confirm the potential in the clinical setting, the present study evaluated the biosafety profile of human MSCs engineered by the hybrid vectors. We unraveled that transduction of MSCs with the hybrid baculovirus vectors slightly impeded the cell proliferation after transduction, probably due to the perturbation of cellular gene expression and induction of innate responses. Nonetheless, the hybrid baculovirus vectors did not compromise the cell viability and cellular differentiation. No transgene integration into the host chromosome and disruption of the karyotype of the MSCs were observed. Additionally, no upregulation of proto-oncogenes or downregulation of tumor suppressor genes occurred in the MSCs transduced with the hybrid baculovirus vectors. Neither did the transduced MSCs induce tumor formation in nude mice. This study not only supported the safety of MSCs for cell therapy but also implicated the potential of the human MSCs engineered by the hybrid baculovirus vectors for their applications in clinical scenarios necessitating sustained transgene expression.


Vaccine | 2010

Baculovirus as an avian influenza vaccine vector: differential immune responses elicited by different vector forms.

Chi-Yuan Chen; Hung-Jen Liu; Ching-Ping Tsai; Cheng-Yu Chung; Yung-Shen Shih; Poa-Chun Chang; Yung-Tsung Chiu; Yu-Chen Hu

Baculovirus is an enveloped virus that infects insects in nature and has emerged as a novel vaccine vector. We previously constructed a recombinant baculovirus displaying the hemagglutinin protein (HA) of avian influenza virus (AIV) on the viral envelope (Bac-HA64), and demonstrated the induction of humoral responses in immunized mice. To improve the vector design and explore how the vector forms influence the vaccine efficacy, we constructed two more baculoviruses Bac-CHA and Bac-CHA/HA64. Bac-CHA expressed HA after transducing the host cells while Bac-CHA/HA64 not only expressed HA but also displayed HA on the envelope. After administration into BALB/c mice, all three vectors elicited HA-specific humoral (IgG1, IgG2a and hemagglutination inhibition titers), mucosal (IgA titers) and cellular (interferon (IFN)-γ and IL-4 producing T cells and IFN-γ(+)/CD8(+) T cells) immune responses. Intriguingly, the magnitudes and types of responses hinged on the vaccine form and administration route. Via intranasal (i.n.) and subcutaneous (s.c.) inoculation, the HA-displaying vectors Bac-HA64 and Bac-CHA/HA64 triggered stronger humoral and mucosal responses than Bac-CHA, but upon intramuscular (i.m.) injection the HA-expressing vectors (Bac-CHA and Bac-CHA/2HA64) elicited more robust humoral and cellular responses than Bac-HA64. Via either administration route, the dual form vaccine Bac-CHA/HA64 gave rise to superior or at least comparable HA-specific immune responses than the other two vaccine forms, implicating the potential of Bac-CHA/HA64 as a vaccine candidate against AIV infection.


Biotechnology Progress | 2009

Concanavalin a affinity chromatography for efficient baculovirus purification

Guan-Yu Chen; Chi-Yuan Chen; Margaret Dah-Tsyr Chang; Yoshiharu Matsuura; Yu-Chen Hu

Baculovirus has emerged as a novel gene delivery and vaccine vector, and the demand for purified baculovirus is rising due to the increasing in vivo applications. Since the baculoviral envelope protein gp64 is a glycoprotein, we aimed to develop a concanavalin A (Con A) chromatography process, which harnessed the possible affinity interaction between gp64 and Con A, for simple and effective baculovirus purification. Throughout the purification process the virus stability and recovery were assessed by quantifying the virus transducing titers [TT, defined as transducing units (TU) per milliliter] and viral particles (VP). We found that baculovirus stability was sensitive to buffer conditions and diafiltration with a tangential flow filtration system LabScale using 300 K membranes yielded recoveries of ≈75% in TT and 82% in VP. The diafiltered baculovirus strongly bound to the Con A column as evidenced by the low virus losses to the flow through and wash fractions. The wash steps eliminated >99% of protein impurities and elution with 0.6 M α‐D‐methylmannoside at room temperature led to the recoveries of ≈16% in VP and ≈15.3% in TU. The resultant VP/TU ratio was as low as 41.4, attesting the high quality of the purified virus. Further elution with 1 M α‐D‐methylmannoside recovered another 6% virus TU, yielding a cumulative recovery of ≈21.3% in TU. These data demonstrated for the first time that Con A chromatography is suitable for baculovirus purification, and may be used for the purification of other viruses with surface glycoproteins.


Solid State Communications | 1993

Electric-field controllable photoluminescence in porous silicon

Juh Tzeng Lue; Kuang Yao Lo; Shaw-Kwen Ma; Chi-Yuan Chen; C.S. Chang

Abstract Intense photoluminescences (PL) at wavelenghts near 600 nm are observed when either the (100) or (111) surface of the electrochemically etched silicon wafers are illuminated by the 514.5 nm argon laser line. A fascinating phenomenon has been discovered indicating that the PL intensity can be suppressed exhaustively by applying an electric field parallel to the surface. The PL recovers its intensity very slowly when the bias is taken off, suggesting that the slow relaxation of the accumulate charges inside the porous silicon.


Journal of Gene Medicine | 2008

Sustained baculovirus-mediated expression in myogenic cells

Heng-Chun Shen; Chia-Ni Yeh; Guan-Yu Chen; Shiu-Feng Huang; Chi-Yuan Chen; Yung-Chung Chiu; Yu-Chen Hu

Baculovirus has emerged as a promising gene delivery vector due to its low cytotoxicity and nonreplication nature in mammalian cells. However, baculovirus‐mediated expression is transient and generally lasts less than 14 days, which could restrict its application in the treatment of diseases requiring stable transgene expression.

Collaboration


Dive into the Chi-Yuan Chen's collaboration.

Top Co-Authors

Avatar

Yu-Chen Hu

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Wen-Hsin Lo

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Guan-Yu Chen

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Chin-Yu Lin

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Cheng-Yu Chung

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Chia-Ni Yeh

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Ching-Kuang Chuang

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Hsin-Yi Chiu

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Shih-Yeh Lin

National Tsing Hua University

View shared research outputs
Top Co-Authors

Avatar

Shiu-Feng Huang

National Health Research Institutes

View shared research outputs
Researchain Logo
Decentralizing Knowledge