Network


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

Hotspot


Dive into the research topics where Leying Zhang is active.

Publication


Featured researches published by Leying Zhang.


NeuroImage | 2007

INTERNALIZATION OF MWCNTS BY MICROGLIA: POSSIBLE APPLICATION IN IMMUNOTHERAPY OF BRAIN TUMORS

Babak Kateb; Michelle Van Handel; Leying Zhang; Michael J. Bronikowski; Harish Manohara; Behnam Badie

There is a pressing need for new therapeutic, diagnostic, and drug delivery approaches for treating brain cancers. Nanotechnology offers a new method for targeted brain cancer therapy and could play a major role in gene and drug delivery. The goals of our study were to visualize in vitro ingestion, cytotoxicity, and loading capacity of Multi-Walled Carbon Nanotubes (MWCNTs) in microglia. Furthermore, we investigated internalization differences between microglia and glioma cells. BV2 microglia and GL261 glioma cells were incubated with MWCNTs, which were synthesized through catalytic chemical vapor deposition technique. Real-time RT-PCR, cell proliferation analysis, siRNA and DNA loading, electron microscopy, and flow cytometry were performed. We demonstrated that MWCNTs do not result in proliferative or cytokine changes in vitro, are capable of carrying DNA and siRNA and are internalized at higher levels in phagocytic cells as compared to tumor cells. This study suggests MWCNTs could be used as a novel, non-toxic, and biodegradable nano-vehicles for targeted therapy in brain cancers. Further studies are needed to demonstrate the full capacity of MWCNTs as nanovectors.


Glia | 2009

Stat3 inhibition activates tumor macrophages and abrogates glioma growth in mice

Leying Zhang; Darya Alizadeh; Michelle Van Handel; Marcin Kortylewski; Hua Yu; Behnam Badie

As the main effector‐cell population of the central nervous system, microglia (MG) are considered to play an important immunoregulatory function in a number of pathological conditions such as inflammation, trauma, degenerative disease, and brain tumors. Recent studies, however, have suggested that the anti‐neoplastic function of MG may be suppressed in malignant brain tumors. Considering the proposed suppressive role of signal transducers and activators of transcription 3 (Stat3) in antitumor immunity, we evaluated the role of Stat3 inhibition on MG and macrophage (MP) activation and tumor growth in a murine glioma model. N9 MG cells were exposed to GL261 glioma conditioned medium (GL261‐CM) and evaluated for Stat3 activity and cytokine expression. Furthermore, the role of Stat3 inhibition on MG and MP activation was studied both in vitro and in vivo. Finally, the effect of Stat3 inhibition on tumor growth was assessed in intracranial GL261 gliomas. GL261‐CM increased Stat3 activity in N9 cells in vitro and resulted in overexpression of IL‐10 and IL‐6, and downregulation of IL1‐β, a pro‐inflammatory cytokine. Inhibition of Stat3 by CPA‐7 or siRNA reversed glioma‐induced cytokine expression profile in N9 cells. Furthermore, inactivation of Stat3 in intracranial GL261 tumors by siRNA resulted in MG/MP activation and tumor growth inhibition. Glioma‐induced MG and MP suppression may be mediated thorough Stat3. Inhibition of Stat3 function in tumor MG/MP may result in their activation and can potentially be used as an adjunct immunotherapy approach for gliomas.


Journal of Neuroimmunology | 2009

Selective uptake of multi-walled carbon nanotubes by tumor macrophages in a murine glioma model

Michelle VanHandel; Darya Alizadeh; Leying Zhang; Babak Kateb; Michael J. Bronikowski; Harish Manohara; Behnam Badie

Carbon nantotubes (CNTs) are emerging as a new family of nanovectors for drug and gene delivery into biological systems. To evaluate potential application of this technology for brain tumor therapy, we studied uptake and toxicity of multi-walled CNTs (MWCNTs) in the GL261 murine intracranial glioma model. Within 24 h of a single intratumoral injection of labeled MWCNTs (5 microg), nearly 10-20% of total cells demonstrated CNT internalization. Most CNT uptake, however, occurred by tumor-associated macrophages (MP), which accounted for most (75%) MWCNT-positive cells. Within 24 h of injection, nearly 30% of tumor MP became MWCNT-positive. Despite a transient increase in inflammatory cell infiltration into both normal and tumor-bearing brains following MWCNT injection, no significant toxicity was noted in mice, and minor changes in tumor cytokine expression were observed. This study suggests that MWCNTs could potentially be used as a novel and non-toxic vehicle for targeting MP in brain tumors.


Nanomedicine: Nanotechnology, Biology and Medicine | 2010

Tumor-associated macrophages are predominant carriers of cyclodextrin-based nanoparticles into gliomas.

Darya Alizadeh; Leying Zhang; Jungyeon Hwang; Thomas Schluep; Behnam Badie

UNLABELLED The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. Using mixed in vitro culture systems, we demonstrated that CDP-NPs were preferentially taken up by BV2 and N9 microglia (MG) cells compared with GL261 glioma cells. Fluorescent microscopy and flow cytometry analysis of intracranial GL261 gliomas confirmed these findings and demonstrated a predominant CDP-NP uptake by macrophages (MPs) and MG within and around the tumor site. Notably, in mice bearing bilateral intracranial tumor, MG and MPs carrying CDP-NPs were able to migrate to the contralateral tumors. In conclusion, these studies better characterize the cellular distribution of CDP-NPs in intracranial tumors and demonstrate that MPs and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors. FROM THE CLINICAL EDITOR The goal of this study was to evaluate the mechanism of cyclodextrin-based nanoparticle (CDP-NP) uptake into a murine glioma model. CDP-NP was preferentially taken up microglia (MG) cells as compared to glioma cells. A predominant CDP-NP uptake by macrophages and MG was also shown in and around the tumor site. Macrophages and MG could potentially be used as nanoparticle drug carriers into malignant brain tumors.


Clinical Cancer Research | 2010

Induction of Anti-Glioma Natural Killer Cell Response following Multiple Low-Dose Intracerebral CpG Therapy

Darya Alizadeh; Leying Zhang; Christine E. Brown; Omar Farrukh; Michael C. Jensen; Behnam Badie

Purpose: Stimulation of toll-like receptor-9 by CpG oligodeoxynucleotides (CpG-ODN) has been shown to counteract the immunosuppressive microenvironment and to inhibit tumor growth in glioma models. These studies, however, have used high doses of CpG-ODN, which can induce toxicity in a clinical setting. The goal of this study was to evaluate the antitumor efficacy of multiple low-dose intratumoral CpG-ODN in a glioma model. Experimental Design: Mice bearing 4-day-old intracranial GL261 gliomas received a single or multiple (two or four) intratumoral injections of CpG-ODN (3 μg) every 4 days. Tumor growth was measured by bioluminescent imaging, brain histology, and animal survival. Flow cytometry and cytotoxicity assays were used to assess anti-glioma immune response. Results: Two and four intracranial injections of low-dose CpG-ODN, but not a single injection, eradicated gliomas in 70% of mice. Moreover, surviving animals exhibited durable tumor-free remission (> 3 months) and were protected from intracranial rechallenge with GL261 gliomas, showing the capacity for long-term antitumor immunity. Although most inflammatory cells seemed to increase, activated natural killer (NK) cells (i.e., NK+CD107a+) were more frequent than CD8+CD107a+ in the brains of rechallenged CpG-ODN–treated animals and showed a stronger in vitro cytotoxicity against GL261 target cells. Leukocyte depletion studies confirmed that NK cells played an important role in the initial CpG-ODN antitumor response, but both CD8 and NK cells were equally important in long-term immunity against gliomas. Conclusions: These findings suggest that multiple low-dose intratumoral injections of CpG-ODN can eradicate intracranial gliomas possibly through mechanisms involving NK-mediated effector function. Clin Cancer Res; 16(13); 3399–408. ©2010 AACR.


Glia | 2011

S100B attenuates microglia activation in gliomas: possible role of STAT3 pathway.

Leying Zhang; Wei Liu; Darya Alizadeh; Dongchang Zhao; Omar Farrukh; Jeffrey Lin; Sam A. Badie; Behnam Badie

Despite significant infiltration into tumors, the effector function of macrophages (MPs) and microglia (MG) appears to be suppressed in gliomas. Although STAT3 pathway is thought to play a role in this process, the exact mechanism by which gliomas induce STAT3 activation in MPs and MG is not known. Because activation of receptor for advanced glycation end products (RAGE) can induce STAT3, and because gliomas express high levels of S100B, a RAGE ligand, we hypothesized that MP/MG STAT3 activity may be modulated through S100B‐RAGE interaction. Exposure of N9 MG and bone marrow‐derived monocytes (BMM) to GL261 glioma condition medium (GCM) and low (nM) levels of S100B increased RAGE expression, induced STAT3 and suppressed MG function in vitro. Furthermore, neutralization of S100B in GCM, partially reversed IL‐1β suppression in BMM, suggesting that the inhibitory effect of GCM to be in part due to S100B. Finally, blockage of S100B‐RAGE interaction inhibited STAT3 activation in N9 MG and in glioma MG/MP in vivo. These findings suggest that the RAGE pathway may play an important role in STAT3 induction in glioma‐associated MG/MPs, and that this process may be mediated through S100B.


Clinical Cancer Research | 2013

S100B Promotes Glioma Growth through Chemoattraction of Myeloid-Derived Macrophages

Huaqing Wang; Leying Zhang; Ian Y. Zhang; Xuebo Chen; Anna Carolina Carvalho da Fonseca; Shihua Wu; Hui Ren; Sam Badie; Sam Sadeghi; Mao Ouyang; Charles Warden; Behnam Badie

Purpose: S100B is member of a multigenic family of Ca2+-binding proteins, which is overexpressed by gliomas. Recently, we showed that low concentrations of S100B attenuated microglia activation through the induction of Stat3. We hypothesized that overexpression of S100B in gliomas could promote tumor growth by modulating the activity of tumor-associated macrophages (TAM). Experimental Design: We stably transfected GL261 glioma cell lines with constructs that overexpressed (S100Bhigh) or underexpressed (S100Blow) S100B and compared their growth characteristics to intracranial wild-type (S100Bwt) tumors. Results: Downregulation of S100B in gliomas had no impact on cell division in vitro but abrogated tumor growth in vivo. Interestingly, compared to S100Blow tumors, S100Bwt and S100Bhigh intracranial gliomas exhibited higher infiltration of TAMs, stronger inflammatory cytokine expression, and increased vascularity. To identify the potential mechanisms involved, the expression of the S100B receptor, receptor for advanced glycation end products (RAGE), was evaluated in gliomas. Although S100B expression induced RAGE in vivo, RAGE ablation in mice did not significantly inhibit TAM infiltration into gliomas, suggesting that other pathways were involved in this process. To evaluate other mechanisms responsible for TAM chemoattraction, we then examined chemokine pathways and found that C-C motif ligand 2 (CCL2) was upregulated in S100Bhigh tumors. Furthermore, analysis of The Cancer Genome Atlass glioma data bank showed a positive correlation between S100B and CCL2 expression in human proneural and neural glioma subtypes, supporting our finding. Conclusions: These observations suggest that S100B promotes glioma growth by TAM chemoattraction through upregulation of CCL2 and introduces the potential utility of S100B inhibitors for glioma therapy. Clin Cancer Res; 19(14); 3764–75. ©2013 AACR.


Clinical Cancer Research | 2012

Intracerebral CpG Immunotherapy with Carbon Nanotubes Abrogates Growth of Subcutaneous Melanomas in Mice

Haitao Fan; Ian Y Zhang; Xuebo Chen; Leying Zhang; Huaqing Wang; Anna Carolina Carvalho da Fonseca; Edwin R. Manuel; Don J. Diamond; Andrew Raubitschek; Behnam Badie

Purpose: Recently, we showed that intratumoral delivery of low-dose, immunostimulatory CpG oligodeoxynucleotides conjugated with carbon nanotubes (CNT-CpG) was more effective than free CpG and not only eradicated intracranial (i.c.) gliomas but also induced antitumor immunity that protected mice from subsequent i.c. or systemic tumor rechallenge. Here, we examined whether the same “intracerebral immunotherapy” strategy could be applied to the treatment of metastatic brain tumors. Experimental Design: Mice with both i.c. and s.c. melanomas were injected intratumorally with CNT-CpG into either location. Antitumor responses were assessed by flow cytometry, bioluminescent imaging, and animal survival. Results: When given s.c., CNT-CpG response was mostly local, and it only modestly inhibited the growth of i.c. melanomas. However, i.c. CNT-CpG abrogated the growth of not only brain but also s.c. tumors. Furthermore, compared with s.c. injections, i.c. CNT-CpG elicited a stronger inflammatory response that resulted in more potent antitumor cytotoxicity and improved in vivo trafficking of effector cells into both i.c. and s.c. tumors. To investigate factors that accounted for these observations, CNT-CpG biodistribution and cellular inflammatory responses were examined in both tumor locations. Intracranial melanomas retained the CNT-CpG particles longer and were infiltrated by Toll-like receptor (TLR-9)–positive microglia. In contrast, myeloid-derived suppressive cells were more abundant in s.c. tumors. Although depletion of these cells before s.c. CNT-CpG therapy enhanced its cytotoxic responses, antitumor responses to brain melanomas were unchanged. Conclusions: These findings suggest that intracerebral CNT-CpG immunotherapy is more effective than systemic therapy in generating antitumor responses that target both brain and systemic melanomas. Clin Cancer Res; 18(20); 5628–38. ©2012 AACR.


Journal of Neuroimmunology | 2007

Regulation of IL-10 expression by upstream stimulating factor (USF-1) in glioma-associated microglia ☆

Leying Zhang; Michelle Van Handel; Jill M. Schartner; Aaron Hagar; Grant Allen; Marjorie Curet; Behnam Badie

Understanding the local CNS immune response to neoplasms is essential in the development of immune-based treatments for malignant brain tumors. Using rodent glioma models, we have recently found tumor-associated microglia/macrophages (MG/MP) to be less responsive to known MG/MP activators such as CpG, LPS and IFN-gamma. To understand the mechanism of MG/MP suppression, nuclear extracts from rodent intracranial C6 gliomas, C6 glioma-associated MG/MP, normal brain, and normal MG/MP were obtained and studied using Electrophoretic Mobility Shift Assay (EMSA). Among the nuclear factors studied (AP-1, IRF, USF-1 and Stat-1) only USF-1, which is constitutively expressed in most cells, was down-regulated in tumor-associated MG/MP, but not normal MG/MP. Because tumor-associated MG/MP had higher expression of IL-10 (but not TNF-alpha or TGF-beta), we evaluated the role of USF-1 on IL-10 expression. siRNA mediated inhibition of USF-1 expression in primary MG/MP cultures resulted in up-regulation of IL-10 mRNA but not TNF-alpha or TGF-beta. These findings suggest that USF-1 may play a role in IL-10 regulation in MG/MP in brain tumors.


Methods of Molecular Biology | 2010

Carbon Nanotube Uptake and Toxicity in the Brain

Leying Zhang; Darya Alizadeh; Behnam Badie

The development of novel drug delivery systems is essential for the improvement of therapeutics for most human diseases. Currently used cellular delivery systems, such as viral vectors, liposomes, cationic lipids, and polymers, may have limited clinical efficacy because of safety issues, low gene transfer efficiency, or cytotoxicity. Carbon nanotubes (CNTs) have garnered much interest as possible biological vectors after the recent discovery of their capacity to penetrate cells. Inspite of the prominence of CNT studies in the nanotechnology literature, exploration of their application to central nervous system (CNS) therapeutics is at a very early stage. Before CNTs are used for treatment of brain and spinal cord disorders, however, several issues such as their CNS penetration and toxicity need to be addressed. Here, we discuss methods by which CNT uptake and toxicity can be assessed in animal models.

Collaboration


Dive into the Leying Zhang's collaboration.

Top Co-Authors

Avatar

Behnam Badie

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Darya Alizadeh

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michelle Van Handel

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Babak Kateb

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dongchang Zhao

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Harish Manohara

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Ian Zhang

City of Hope National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Michael J. Bronikowski

California Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Omar Farrukh

City of Hope National Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge