Network


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

Hotspot


Dive into the research topics where Shi-Cheng Zhao is active.

Publication


Featured researches published by Shi-Cheng Zhao.


Human Gene Therapy | 1999

Expression of a novel double-mutant dihydrofolate reductase-cytidine deaminase fusion gene confers resistance to both methotrexate and cytosine arabinoside.

Axel Sauerbrey; Shi-Cheng Zhao; Debabrata Banerjee; Joseph R. Bertino

A novel fusion gene consisting of the open reading frame of a double-mutant (Phe22-Ser31) dihydrofolate reductase (dmDHFR) cDNA fused to the open reading frame of cytidine deaminase (CD) was constructed and characterized for the purpose of conferring simultaneous resistance to methotrexate (MTX) and cytosine arabinoside (ara-C). The kinetic properties of purified recombinant dmDHFR-CD fusion protein were compared with those of purified CD and dmDHFR. The fusion protein was found to retain enzymatic properties of both dmDHFR and CD, in that the Km and Kcat values of purified dmDHFR-CD protein were found to be virtually identical to those of CD and dmDHFR alone. Retrovirus-mediated expression of dmDHFR-CD in NIH 3T3 cells conferred significant resistance (10- to 12-fold) against MTX and ara-C, compared with mock- and single gene-infected cells and the level of resistance obtained was similar to that of cells expressing both CD and dmDHFR from a retroviral bicistronic vector. Infection of mouse bone marrow cells with the dmDHFR-CD construct also showed high levels of resistance to MTX and ara-C in a CFU-GM assay. This fusion protein confers resistance to two antineoplastic agents that differ in their mechanism of action, and may be useful in the design of gene transfer strategies for protection of target cells against multiple drugs. Since high-dose ara-C and MTX are used in the treatment of lymphomas, this vector may be of value in protecting human hematopoietic progenitor cells from the toxicity of these antimetabolites.


Journal of Biological Chemistry | 1998

Probing the folate-binding site of human thymidylate synthase by site-directed mutagenesis. Generation of mutants that confer resistance to raltitrexed, Thymitaq, and BW1843U89.

Youzhi Tong; Xinyue Liu-Chen; Emine A. Ercikan-Abali; Shi-Cheng Zhao; Debabrata Banerjee; Frank Maley; Joseph R. Bertino

Human thymidylate synthase (TS) contains three highly conserved residues Ile-108, Leu-221, and Phe-225 that have been suggested to be important for cofactor and antifolate binding. To elucidate the role of these residues and generate drug-resistant human TS mutants, 14 variants with multiple substitutions of these three hydrophobic residues were created by site-directed mutagenesis and transfected into mouse TS-negative cells for complementation assays and cytotoxicity studies, and the mutant proteins expressed and characterized. The I108A mutant confers resistance to raltitrexed and Thymitaq with respective IC50 values 54- and 80-fold greater than wild-type but less resistance to BW1843U89 (6-fold). The F225W mutant displays resistance to BW1843U89 (17-fold increase in IC50 values), but no resistance to raltitrexed and Thymitaq. It also confers 8-fold resistance to fluorodeoxyuridine. Both the kinetic characterization of the altered enzymes and formation of antifolate-resistant colonies in mouse bone marrow cells that express mutant TS are in accord with the IC50 values for cytotoxicity noted above. The human TS mutants (I108A and F225W), by virtue of their desirable properties, including good catalytic function and resistance to antifolate TS inhibitors, confirm the importance of amino acid residues Ile-108 and Phe-225 in the binding of folate and its analogues. These novel mutants may be useful for gene transfer experiments to protect hematopoietic progenitor cells from the toxic effects of these drugs.


Gene | 1994

Transfection with a cDNA encoding a Ser31 or Ser35 mutant human dihydrofolate reductase into Chinese hamster ovary and mouse marrow progenitor cells confers methotrexate resistance

Debabrata Banerjee; Barry Schweitzer; Matthias Volkenandt; Ming-Xia Li; Mark Waltham; Shin Mineishi; Shi-Cheng Zhao; Joseph R. Bertino

Chinese hamster ovary (CHO) DHFR- cells were converted into the DHFR+ phenotype when they were transfected with a mammalian expression vector carrying human dihydrofolate reductase-encoding cDNAs (DHFR) containing a Ser31 or a Ser34 mutation. Furthermore, transfection of these mutants into wild-type CHO cells resulted in resistance to high levels of methotrexate (MTX), indicating that these human variants can act as dominant selectable markers. Southern blot analysis and polymerase chain reaction amplifications confirmed that the transfected plasmids were integrated into the CHO DNA. Gene copy number analysis revealed that both the Ser3 1 and the Ser3.4 mutants amplifiable when grown in increasing concentrations of MTX. Retrovirus-mediated gene transfer of the Ser31 mutant into mouse marrow progenitor cells also resulted in MTX-resistant CFU-GM (colony-forming unit-granulocyte macrophage) cells.


Gene Therapy | 1997

Co-expression of the herpes simplex virus thymidine kinase gene potentiates methotrexate resistance conferred by transfer of a mutated dihydrofolate reductase gene

Shin Mineishi; S. Nakahara; Naoko Takebe; Debabrata Banerjee; Shi-Cheng Zhao; Joseph R. Bertino

We have previously shown that transfer of a mutated dihydrofolate reductase (DHFR) confers resistance to methotrexate (MTX) to infected cells. We report herein the construction of a retrovirus vector, DC/SV6S31tk, which carries the herpes simplex virus thymidine kinase gene (HSVtk) as well as the mutated Serine 31 DHFR (S31) cDNA. 3T3 cells infected with DC/SV6S31tk are more resistant to MTX than cells infected with DC/SV6S31, which carries the S31 and Neor gene. In DC/SV6S31tk-infected cells, a fraction of cells (20–40%) were more resistant to MTX compared with DC/SV6S31-infected cells, and these cells survived a 5-day exposure to 200 μ M of MTX. The mechanism of this augmented resistance is attributed to the salvage of thymidine by HSVtk, as the augmentation is reversed when dialyzed serum is used for cytotoxicity assays. The cells that survive high-dose MTX selection have high levels of expression of S31 DHFR and HSVtk, although copy numbers of the proviral sequences do not increase significantly. Transduction of cells with the DC/SV6S31tk vector also sensitizes cells to ganciclovir (GCV). Co-expression of a metabolically related gene in a retroviral vector to potentiate the resistance imparted by a drug resistance gene may be useful for gene therapy for cancer patients.


Cancer Gene Therapy | 2000

Comparison of methotrexate resistance conferred by a mutated dihydrofolate reductase (DHFR) cDNA in two different retroviral vectors

Naoko Takebe; Saori Nakahara; Shi-Cheng Zhao; Debasis Adhikari; Ali Ugur Ural; Marian Iwamoto; Debabrata Banerjee; Joseph R. Bertino

We previously reported the protection of hematopoietic cells from methotrexate (MTX) toxicity using an N2-based double copy vector containing serine 31 (S31)-mutated dihydrofolate reductase (DHFR) (DC/SV6S31). To examine whether the use of SFG-based dicistronic vectors will lead to improvement in gene transfer over the DC/SV6 vector, we compared the protection provided by MTX to NIH3T3 cells and hematopoietic progenitor cells infected with these retroviral constructs containing the S31 variant DHFR cDNA. In NIH3T3 cells, the 50% effective dose values of MTX conferred by the SFG vector were 8-fold higher than those obtained with the DC/SV6 vector. DHFR mRNA levels were 22-fold and 38-fold higher than that seen for the DC/SV6 vector according to Northern blot and real-time polymerase chain reaction analysis, respectively. However, DHFR protein expression and DHFR enzyme activity were only 1.5-fold and 2-fold higher in the SFG vector, respectively, indicating that the mRNA from the SFG vector is translated less efficiently than the mRNA generated from the DC/SV6 vector. Furthermore, the degree of MTX protection conferred by each vector in both mouse and human hematopoietic cells was the same. These results indicate that the in vitro transduction efficiency and transgene expression of human DHFR in hematopoietic progenitor cells is equally conferred by both vectors.


Cancer Research | 1998

Role of E2F-1 in Chemosensitivity

Debabrata Banerjee; Barbara Schnieders; Jenny Z. Fu; Debasis Adhikari; Shi-Cheng Zhao; Joseph R. Bertino


Cancer Gene Therapy | 1994

Long-term protection of recipient mice from lethal doses of methotrexate by marrow infected with a double-copy vector retrovirus containing a mutant dihydrofolate reductase.

Shi-Cheng Zhao; Ming-Xia Li; Debabrata Banerjee; Barry Schweitzer; Shin Mineishi; Eli Gilboa; Bertino


Molecular Therapy | 2001

Generation of dual resistance to 4-hydroperoxycyclophosphamide and methotrexate by retroviral transfer of the human aldehyde dehydrogenase class 1 gene and a mutated dihydrofolate reductase gene.

Naoko Takebe; Shi-Cheng Zhao; Debasis Adhikari; Shin Mineishi; Michel Sadelain; John Hilton; Michael Colvin; Debabrata Banerjee; Joseph R. Bertino


Journal of Biological Chemistry | 1998

Isolation and Characterization of Thymitaq (AG337) and 5-Fluoro-2-deoxyuridylate-resistant Mutants of Human Thymidylate Synthase from Ethyl Methanesulfonate-exposed Human Sarcoma HT1080 Cells

Youzhi Tong; Xinyue Liu-Chen; Emine A. Ercikan-Abali; Gina M. Capiaux; Shi-Cheng Zhao; Debabrata Banerjee; Joseph R. Bertino


Human Gene Therapy | 1997

Post-Transplant Methotrexate Administration Leads to Improved Curability of Mice Bearing a Mammary Tumor Transplanted with Marrow Transduced with a Mutant Human Dihydrofolate Reductase cDNA

Shi-Cheng Zhao; Debabrata Banerjee; Shin Mineishi; Joseph R. Bertino

Collaboration


Dive into the Shi-Cheng Zhao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Joseph R. Bertino

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Shin Mineishi

Penn State Cancer Institute

View shared research outputs
Top Co-Authors

Avatar

Naoko Takebe

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Bertino

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Debasis Adhikari

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Emine A. Ercikan-Abali

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ming-Xia Li

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge