Network


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

Hotspot


Dive into the research topics where Shangzhen Zhou is active.

Publication


Featured researches published by Shangzhen Zhou.


Nature Medicine | 1999

Human fibroblast growth factor receptor 1 is a co-receptor for infection by adeno-associated virus 2

Arun Srivastava; Keyun Qing; Cathryn Mah; Jonathan J. Hansen; Shangzhen Zhou; Varavani Dwarki

Adeno-associated virus 2 (AAV)-based vectors have gained attention as a potentially useful alternative to the more commonly used retroviral and adenoviral vectors for human gene therapy. Although AAV uses the ubiquitously expressed cell surface heparan sulfate proteoglycan (HSPG) as a receptor, the transduction efficiency of AAV vectors varies greatly in different cells and tissues in vitro and in vivo. We demonstrate here that cell surface expression of HSPG alone is insufficient for AAV infection, and that AAV also requires human fibroblast growth factor receptor 1 (FGFR1) as a co-receptor for successful viral entry into the host cell. We document that cells that do not express either HSPG or FGFR1 fail to bind AAV and, consequently, are resistant to infection by AAV. These non-permissive cells are successfully transduced by AAV vectors after stable transfections with cDNAs encoding the murine HSPG and the human FGFR1. Furthermore, AAV infection of permissive cells, known to express both FGFR1 and the epidermal growth factor receptor, is abrogated by treatment of cells with basic fibroblast growth factor, but not with epidermal growth factor. The identification of FGFR1 as a co-receptor for AAV should provide new insights not only into its role in the life cycle of AAV, but also in the optimal use of AAV vectors in human gene therapy.


Journal of Virology | 2001

Adeno-Associated Virus Type 2-Mediated Gene Transfer: Role of Cellular FKBP52 Protein in Transgene Expression

Keyun Qing; Jonathan J. Hansen; Kirsten A. Weigel-Kelley; Mengqun Tan; Shangzhen Zhou; Arun Srivastava

ABSTRACT Although adeno-associated virus type 2 (AAV) has gained attention as a potentially useful vector for human gene therapy, the transduction efficiencies of AAV vectors vary greatly in different cells and tissues in vitro and in vivo. We have documented that a cellular tyrosine phosphoprotein, designated the single-stranded D-sequence-binding protein (ssD-BP), plays a crucial role in AAV-mediated transgene expression (K. Y. Qing, X.-S. Wang, D. M. Kube, S. Ponnazhagan, A. Bajpai, and A. Srivastava, Proc. Natl. Acad. Sci. USA 94:10879–10884, 1997). We have documented a strong correlation between the phosphorylation state of ssD-BP and AAV transduction efficiency in vitro as well as in vivo (K. Y. Qing, B. Khuntrirat, C. Mah, D. M. Kube, X.-S. Wang, S. Ponnazhagan, S. Z. Zhou, V. J. Dwarki, M. C. Yoder, and A. Srivastava, J. Virol. 72:1593–1599, 1998). We have also established that the ssD-BP is phosphorylated by epidermal growth factor receptor protein tyrosine kinase and that the tyrosine-phosphorylated form, but not the dephosphorylated form, of ssD-BP prevents AAV second-strand DNA synthesis and, consequently, results in a significant inhibition of AAV-mediated transgene expression (C. Mah, K. Y. Qing, B. Khuntrirat, S. Ponnazhagan, X.-S. Wang, D. M. Kube, M. C. Yoder, and A. Srivastava, J. Virol. 72:9835–9841, 1998). Here, we report that a partial amino acid sequence of ssD-BP purified from HeLa cells is identical to a portion of a cellular protein that binds the immunosuppressant drug FK506, termed the FK506-binding protein 52 (FKBP52). FKBP52 was purified by using a prokaryotic expression plasmid containing the human cDNA. The purified protein could be phosphorylated at both tyrosine and serine or threonine residues, and only the phosphorylated forms of FKBP52 were shown to interact with the AAV single-stranded D-sequence probe. Furthermore, in in vitro DNA replication assays, tyrosine-phosphorylated FKBP52 inhibited AAV second-strand DNA synthesis by greater than 90%. Serine- or threonine-phosphorylated FKBP52 caused ≈40% inhibition, whereas dephosphorylated FKBP52 had no effect on AAV second-strand DNA synthesis. Deliberate overexpression of FKBP52 effectively reduced the extent of tyrosine phosphorylation of the protein, resulting in a significant increase in AAV-mediated transgene expression in human and murine cell lines. These studies corroborate the idea that the phosphorylation status of the cellular FKBP52 protein correlates strongly with AAV transduction efficiency, which may have important implications for the optimal use of AAV vectors in human gene therapy.


Gene Therapy | 1998

Adeno-associated virus-mediated delivery of erythropoietin leads to sustained elevation of hematocrit in nonhuman primates.

Shangzhen Zhou; John E. Murphy; Jaime Escobedo; Varavani Dwarki

Recombinant adeno-associated virus encoding the monkey erythropoietin gene (rAAV-cm-Epo) was generated and tested for its potential to confer long-term expression of the gene product following intramuscular injection. A single intramuscular injection of 2 × 1012 rAAV-cm-Epo particles into two baboons led to sustained high circulating Epo levels and a concomitant increase in hematocrit. The hematocrits reached 62 and 75% by week 10 (from pre- injection values of 38 and 40%, respectively) and remained elevated throughout the study period (28 weeks). Circulating Epo levels were also elevated throughout the study period. Our data demonstrate the potential for long-term gene expression in large animals by a single intramuscular injection of a recombinant adeno-associated virus (rAAV) vector.


Virology | 1992

Parvovirus B19 replication in human umbilical cord blood cells

Carolyn H. Srivastava; Shangzhen Zhou; Nikhil C. Munshi; Arun Srivastava

The human parvovirus B19 is now known to be one of the causative agents of nonimmune hydrops fetalis and spontaneous abortions in pregnant women. The presence of the viral proteins and antibodies in fetuses of B19-infected women suggests that the virus can cross the placental barrier. In order to gain an insight into the mechanism of intrauterine fetal infection and the virus-induced hydrops fetalis, we examined whether human umbilical cord blood cells were permissive for B19 replication. Cord blood cells were infected with B19 in vitro, and Southern blot analyses of low M(r) DNA isolated from these cells revealed the presence of the characteristic replicative intermediates of B19 DNA. In addition, B19 genome expression in cord blood cells was detected by Northern blot analysis. Quantitative DNA dot blot analysis of culture supernatants documented complete assembly and release of B19 progeny virions in these cells. The progeny virions were biologically active in secondary infections of normal human bone marrow cells. The human umbilical cord blood cells may be a useful alternative to bone marrow and fetal liver culture systems for further studies on B19 since the need for bone marrow donors is obviated and, unlike fetal tissues, there are no ethical questions associated with the experimental use of cord blood because it is normally discarded. These studies also suggest that the umbilical cord blood may be a site for active replication of parvovirus B19 in vivo and may thus provide a means for transmission of the virus during intrauterine fetal infections.


Gene Therapy | 2006

Enhanced transduction of mouse bone marrow-derived dendritic cells by repetitive infection with self-complementary adeno-associated virus 6 combined with immunostimulatory ligands

Wayne Aldrich; Changchun Ren; White Af; Shangzhen Zhou; Sanjay Kumar; Connie Jenkins; Denise R. Shaw; Theresa V. Strong; Pierre L. Triozzi; Selvarangan Ponnazhagan

The potential of adeno-associated virus (AAV)-based vectors in human gene therapy is being explored for several diseases. Although sustained transgene expression and low vector-associated cellular immunity are attractive features of recombinant (r) AAV, the wider application of rAAV vectors encapsidated in serotype 2 capsid is hampered by poor transduction efficiency in many target tissues. These include ex vivo-generated dendritic cells (DC), which have demonstrated promising immunotherapeutic activity. We report here that efficient transduction of mouse bone marrow-derived DC can be achieved with self-complementary (sc) rAAV encapsidated in serotype 6 capsid. Sequential exposure of DC precursor cultures to IL-4 and GM-CSF with sc rAAV6 encoding the human tumor antigen, carcinoembryonic antigen (CEA), for 7 days followed by activation with CpG oligodeoxynucleotides (ODN) and anti-mouse CD40 antibody resulted in highly efficient transduction of DC. DC surface markers as determined by flow cytometry analysis of sc rAAV6-transduced DC were comparable to nontransduced DC. Efficiency of vector transduction and transgene expression were confirmed by immunostaining and real-time PCR. Microarray analysis of RNA from CpG ODN and CD40 antibody stimulated sc AAV6-transduced DC revealed upregulation of transcription factors and cytokines involved in immune activation and downregulation of inhibitory factors, suggesting a possible role of transcriptional activation in the observed effect. The adoptive transfer into syngeneic mice of the ex vivo-transduced and activated DC resulted in the development of CEA-specific antibody and T-helper 1-associated immune responses. Immunized mice also developed antibody to AAV6 capsid protein, which did not crossreact with AAV2 capsid protein. These studies demonstrate the potential utility of sc rAAV serotype 6-based vectors in transduction of DC for genetic vaccination approaches.


Current Topics in Microbiology and Immunology | 1996

Adeno-associated Virus 2-Mediated Transduction and Erythroid Lineage-Specific Expression in Human Hematopoietic Progenitor Cells

Arun Srivastava; Xu-Shan Wang; Selvarangan Ponnazhagan; Shangzhen Zhou; Mervin C. Yoder

Parvoviruses are among the smallest of the DNA-containing viruses that infect a wide variety of vertebrates (Siegl et al. 1985). Two parvoviruses of human origin, the nonpathogenic adeno-associated virus 2 (AAV) and the parvovirus B19, a common human pathogen, have been studied extensively (Berns and Bohenzky 1987; Brown et al. 1994). AAV requires coinfection with a helper virus, such as adenovirus or herpesvirus, for its optimal replication (Berns 1990), but in the absence of a helper virus, the AAV genome establishes a latent infection in a site-specific manner (Kotin and Berns 1989; Kotin et al. 1990, 1991, 1992; Samulski et al. 1991). B19, by contrast, is an autonomously replicating virus with a remarkable tropism for human erythroid progenitor cells (Ozawa et al. 1986, 1987; Yaegashi et al. 1989; Srivastava and Lu 1988; Takahashi et al. 1990). We have described the construction of a recombinant AAV-B19 hybrid genome, in which we combined the remarkable features of these two parvoviruses, and speculated that such a hybrid vector may prove useful for high efficiency transduction of primary human hematopoietic progenitor cells (Srivastava et al. 1989). Indeed, it has become increasingly clear that the AAV-based vector system may prove to be a useful alternative to the more commonly used retroviral and adenoviral vectors for its potential use in human gene therapy (Muzyczka 1992; Carter 1993; Srivastava 1994). Despite these advances, a number of fundamental questions related to AAV remain unanswered. For example, the molecular details of viral assembly and the mechanism of viral entry into the host cell have not been rigorously analyzed. Furthermore, the feasibility of obtaining tissue-specific expression of an AAV-transduced gene has not been adequately addressed. Here, we provide experimental evidence to suggest that the vector assembly requires a precise signaling mechanism and that AAV infection of human cells is receptor-mediated. We also document erythroid lineage restricted expression following AAV-B19 hybrid vector-mediated transduction of primary human hematopoietic progenitor cells. Elucidation of the molecular details of these aspects of AAV biology will have important implications in the potential use of AAV as a vector in human gene therapy.


Molecular Therapy | 2005

849. Transduction of Mouse Dendritic Cells Is Enhanced by Repetitive Infection with Self-Complementary Adeno-Associated Virus 6 Combined with Immunostimulatory Ligands

Wayne A. Aldrich; Changchun Ren; AprilF. White; Shangzhen Zhou; Sanjay Kumar; Connie Jenkins; Denise R. Shaw; Theresa V. Strong; Pierre L. Triozzi; Selvarangan Ponnazhagan

The potential of adeno-associated virus (AAV)-based vectors in human gene therapy is being explored for several diseases. Although sustained transgene expression and low vector-associated cellular immunity are attractive features of recombinant (r) AAV, the wider application rAAV vectors encapsidated in serotype 2 capsid, the most commonly applied rAVV, is hampered by poor transduction efficiency in many target tissues. These include ex vivo generated dendritic cells (DC), which have demonstrated promising immunotherapeutic activity. We report here that efficient transduction of mouse bone marrow-derived DC can be achieved with self-complementary (sc) rAAV encapsidated in serotype 6 capsid. Sequential exposure of DC precursor cultures to IL-4 and GM-CSF with sc rAAV6 encoding the human tumor antigen, carcinoembryonic antigen (CEA), for seven days followed by activation with CpG oligodeoxynucleotides (ODN) and anti-mouse CD40 antibody resulted in highly efficient transduction of DC. DC surface markers as determined by flow cytometry analysis of sc rAAV6-transduced DC were comparable to non-transduced DC. Efficiency of vector transduction and transgene expression were confirmed by immunostaining and real-time PCR. Microarray analysis of RNA from CpG-ODN and CD40 antibody stimulated sc AAV6-transduced DC revealed upregulation of transcription factors and cytokines involved in immune activation and down-regulation of inhibitory factors, suggesting a possible role of transcriptional activation in the observed effect. The adoptive transfer into syngeneic mice of the ex vivo transduced and activated DC resulted in the development of CEA-specific antibody. Immunized mice also developed antibody to AAV-6 capsid protein which did not cross-react with AAV-2 capsid protein. These studies demonstrate the potential utility of sc rAAV serotype 6-based vectors in transduction of DC for genetic vaccination approaches.


Investigative Ophthalmology & Visual Science | 2000

Retinal degeneration is slowed in transgenic rats by AAV-mediated delivery of FGF-2.

Dana Lau; Laura H. Mcgee; Shangzhen Zhou; Katherine Rendahl; William C. Manning; Jaime Escobedo; John G. Flannery


Human Gene Therapy | 1998

Transient Immunosuppression Allows Transgene Expression Following Readministration of Adeno-Associated Viral Vectors

William C. Manning; Shangzhen Zhou; Mary Pat Bland; Jaime Escobedo; Varavani Dwarki


Proceedings of the National Academy of Sciences of the United States of America | 1997

Long-term correction of obesity and diabetes in genetically obese mice by a single intramuscular injection of recombinant adeno-associated virus encoding mouse leptin

John E. Murphy; Shangzhen Zhou; Klaus Giese; Lewis T. Williams; Jaime Escobedo; Varavani Dwarki

Collaboration


Dive into the Shangzhen Zhou's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jaime Escobedo

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge