Network


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

Hotspot


Dive into the research topics where Zheng Ao is active.

Publication


Featured researches published by Zheng Ao.


Cancer Research | 2015

Identification of cancer associated fibroblasts in circulating blood from patients with metastatic breast cancer

Zheng Ao; Sanket H. Shah; Leah Machlin; Ritesh Parajuli; Philip Miller; Siddarth Rawal; Anthony Williams; Richard J. Cote; Marc E. Lippman; Ram H. Datar; Dorraya El-Ashry

Metastasis is facilitated by cancer-associated fibroblasts (CAF) in the tumor microenvironment through mechanisms yet to be elucidated. In this study, we used a size-based microfilter technology developed by our group to examine whether circulating CAF identified by FAP and α-SMA co-expression (cCAF) could be distinguished in the peripheral blood of patients with metastatic breast cancer. In a pilot study of patients with breast cancer, we detected the presence of cCAFs in 30/34 (88%) patients with metastatic disease (MET group) and in 3/13 (23%) patients with localized breast cancer (LOC group) with long-term disease-free survival. No cCAFs as defined were detected in healthy donors. Further, both cCAF and circulating tumor cells (CTC) were significantly greater in the MET group compared with the LOC group. Thus, the presence of cCAF was associated with clinical metastasis, suggesting that cCAF may complement CTC as a clinically relevant biomarker in metastatic breast cancer.


Breast Cancer Research and Treatment | 2014

Primary breast tumor-derived cellular models: characterization of tumorigenic, metastatic, and cancer-associated fibroblasts in dissociated tumor (DT) cultures

Katherine Drews-Elger; Joeli Brinkman; Philip Miller; Sanket H. Shah; J. Chuck Harrell; Thiago G. da Silva; Zheng Ao; Amy Schlater; Diana J. Azzam; Kathleen M. Diehl; Dafydd G. Thomas; Joyce M. Slingerland; Charles M. Perou; Marc E. Lippman; Dorraya El-Ashry

Our goal was to establish primary cultures from dissociation of breast tumors in order to provide cellular models that may better recapitulate breast cancer pathogenesis and the metastatic process. Here, we report the characterization of six cellular models derived from the dissociation of primary breast tumor specimens, referred to as “dissociated tumor (DT) cells.” In vitro, DT cells were characterized by proliferation assays, colony formation assays, protein, and gene expression profiling, including PAM50 predictor analysis. In vivo, tumorigenic and metastatic potential of DT cultures was assessed in NOD/SCID and NSG mice. These cellular models differ from recently developed patient-derived xenograft models in that they can be used for both in vitro and in vivo studies. PAM50 predictor analysis showed DT cultures similar to their paired primary tumor and as belonging to the basal and Her2-enriched subtypes. In vivo, three DT cultures are tumorigenic in NOD/SCID and NSG mice, and one of these is metastatic to lymph nodes and lung after orthotopic inoculation into the mammary fat pad, without excision of the primary tumor. Three DT cultures comprised of cancer-associated fibroblasts (CAFs) were isolated from luminal A, Her2-enriched, and basal primary tumors. Among the DT cells are those that are tumorigenic and metastatic in immunosuppressed mice, offering novel cellular models of ER-negative breast cancer subtypes. A group of CAFs provide tumor subtype-specific components of the tumor microenvironment (TME). Altogether, these DT cultures provide closer-to-primary cellular models for the study of breast cancer pathogenesis, metastasis, and TME.


Cancer Biology & Therapy | 2015

Hierarchical paracrine interaction of breast cancer associated fibroblasts with cancer cells via hMAPK-microRNAs to drive ER-negative breast cancer phenotype

Sanket H. Shah; Philip Miller; Marta Garcia-Contreras; Zheng Ao; Leah Machlin; Emilio Issa; Dorraya El-Ashry

Multiple juxtacrine and paracrine interactions occur between cancer cells and non-cancer cells of the tumor microenvironment (TME) that direct tumor progression. Cancer Associated Fibroblasts (CAFs) are an integral component of the TME, and the majority of breast tumor stroma is comprised of CAFs. Heterotypic interactions between cancer cells and non-cancer cells of the TME occur via soluble agents, including cytokines, hormones, growth factors, and secreted microRNAs. We previously identified a microRNA signature indicative of hyperactive MAPK signaling (hMAPK-miRNA signature) that significantly associated with reduced recurrence-free and overall survival. Here we report that the hMAPK-miRNA signature associates with a high metric of stromal cell infiltrate, and we investigate the role of microRNAs, particularly hMAPK-microRNAs, secreted by CAFs on estrogen receptor (ER) expression in breast cancer cells. ER-positive MCF-7/ltE2- cells were treated with conditioned media (CM) from CAFs derived from breast cancers of different PAM50 subtypes (CAFBAS, CAFHER2, and CAFLA). CAF CM isolated specifically from ER-negative primary breast tumors led to ER repression in vitro. Nanoparticle tracking analysis and transmission electron microscopy confirmed the presence of CAF-secreted exosomes in CM and the uptake of these exosomes by the ER+ MCF-7/ltE2- cells. Differentially expressed microRNAs in CAF CM as well as in MCF-7/ltE2- cells treated with this CM were identified. Knockdown of miR-221/222 in CAFBAS resulted in knockdown of miR221/222 levels in the conditioned media and the CM from CAFBAS; miR221/222 knockdown rescued ER repression in ER-positive cell lines treated with CAFBAS-CM. Collectively, our results demonstrate that CAF-secreted microRNAs are directly involved in ER-repression, and may contribute to the MAPK-induced ER repression in breast cancer cells.


Lab on a Chip | 2016

A surface acoustic wave biosensor for interrogation of single tumour cells in microcavities

Sukru U. Senveli; Zheng Ao; Siddarth Rawal; Ram H. Datar; Richard J. Cote; Onur Tigli

In this study, biological cells are sensed and characterized with surface acoustic wave (SAW) devices utilising microcavities. After tumour cells in media are transported to and trapped in microcavities, the proposed platform uses SAW interaction between the substrate and the cells to extract their mechanical stiffness based on the ultrasound velocity. Finite element method (FEM) analysis and experimental results show that output phase information is an indicator of the stiffness modulus of the trapped cells. Small populations of various types of cells such as MCF7, MDA-MB-231, SKBR3, and JJ012 were characterized and characteristic moduli were estimated for each cell population. Results show that high frequency stiffness modulus is a possible biomarker for aggressiveness of the tumour and that microcavity coupled SAW devices are a good candidate for non-invasive interrogation of single cells.


Lab on a Chip | 2015

Thermoresponsive release of viable microfiltrated Circulating Tumor Cells (CTCs) for precision medicine applications

Zheng Ao; Erika Parasido; Siddarth Rawal; Anthony Williams; Richard Schlegel; Stephen V. Liu; Chris Albanese; Richard J. Cote; Ashutosh Agarwal; Ram H. Datar

Stimulus responsive release of Circulating Tumor Cells (CTCs), with high recovery rates from their capture platform, is highly desirable for off-chip analyses. Here, we present a temperature responsive polymer coating method to achieve both release as well as culture of viable CTCs captured from patient blood samples.


Archive | 2016

Affinity-Based Enrichment of Circulating Tumor Cells

Zheng Ao; Richard J. Cote; Ram H. Datar

Study of CTC in cancer has always been hampered by its rare existence in blood. In this chapter, we discuss one of first principles employed to capture CTC from cancer patients’ peripheral blood—the affinity-based enrichment of CTC. We briefly discuss the different technologies utilizing antibodies to capture CTC based on specific antigen expression. Then we address the downstream molecular and functional characterization of CTC by utilizing these technologies. We also discuss the limitations of affinity-based CTC enrichment.


Archive | 2016

Size-Based and Non-Affinity Based Microfluidic Devices for Circulating Tumor Cell Enrichment and Characterization

Zheng Ao; Kamran Moradi; Richard J. Cote; Ram H. Datar

Circulating Tumor Cells (CTCs) are tumor cells found in cancer patients’ peripheral blood. Enumeration of CTCs can provide prognosis information for cancer management (Cristofanilli et al., N Engl J Med 351(8):781–791, 2004; Cohen et al., J Clin Oncol 26(19):3213–3221, 2008; de Bono et al., Clin Cancer Res, 14(19):6302–6309, 2008; Poveda et al., Gynecol Oncol, 122(3):567–572, 2011). However, the technical hurdle for studying CTCs is their rare presence in blood, thus, isolating them is a non-trivial task. Two major categories of technologies have been developed in the past to isolate CTCs based on their biological expression of antigens (affinity-based capture) or based on their physical properties (non-affinity based capture). This chapter dedicates itself to the non-affinity based method for CTC capture. CTCs, as tumor cells, are inherently distinct from normal blood components. The chapter touches on the how these differences are reflected in their gene expression profiles, as well as their physical properties. We discuss how researchers utilized the unique biomechanical and electrical properties of CTCs to isolate them from enormous numbers of erythrocytes and leukocytes present in peripheral blood. We begin the chapter with technologies utilizing biomechanical properties (cell density, size, deformability) to isolate CTCs and then move on to discuss the development of dielectrophoresis (DEP) based CTC isolation, based on their distinct electrical properties.


2013 IEEE Point-of-Care Healthcare Technologies (PHT) | 2013

Clinical translation of a novel microfilter technology Capture, characterization and culture of circulating tumor cells

Anthony Williams; Siddarth Rawal; Zheng Ao; Jorge Torres-Munoz; Marija Balic; Ming-Da Zhou; Siyang Zheng; Yu-Chong Tai; Richard J. Cote; Ram H. Datar

The most important determinant of prognosis and management of cancer is the presence or absence of metastasis [1]. The road to metastasis involves tumor cells to become detached from the primary tumor and travel in the blood to distant sites, causing secondary tumors. These tumor cells traveling in blood are termed Circulating tumor cells (CTC). Capture of CTC from whole blood has been a challenging feat. The fact that these CTC are few in number, to effectively and efficiently isolate them from whole blood can be thought of as looking for a needle in a haystack. Our microfilter technology exploits the use of size based capture of the larger CTC from the smaller white blood cells and components of whole blood. The effective capture potential of the microfilter platform has driven the area of CTC analysis into a new age of research in the field of cancer. The ability to finally analyze CTC at a molecular level, leads to a deeper understanding of metastatic process, while providing an opportunity to evaluate, monitor and manage treatment options as well as the adherent possibility of having an “on-chip” drug sensitivity assay for focused treatment options. We have demonstrated through clinical trials the ability to effectively identify, enumerate and characterize CTC based on immunfluorescence and FISH assays and provide a companion endpoint for monitoring and evaluating treatment management. Our work on viable CTC capture has resulted in successfully capturing and culturing CTC from blood in mouse models that have been inoculated with breast cancer cell lines to form primary and secondary metastatic cancer sites. The future potential within the microfilter technology to capture viable CTC for culture, will catapult therapeutic interventions to a new level of personalized medicine in cancer management.


Archive | 2016

Significance of Studying Circulating Tumor Cells

Ram H. Datar; Zheng Ao; Richard J. Cote

Circulating Tumor Cells (CTC) are tumor cells released into blood. They are considered the pivotal component of the metastatic cascade and are being extensively studied only in the last decade or so. Understanding the biological and clinical impact of CTC is likely to reveal important information of the metastatic process and contribute to better management of cancer. We briefly discuss here the current clinical utility of CTC and their emerging clinical applications.


Journal of Visualized Experiments | 2016

Capture and Release of Viable Circulating Tumor Cells from Blood

Siddarth Rawal; Zheng Ao; Ashutosh Agarwal

We demonstrate a method for size based capture of viable circulating tumor cell (CTC) from whole blood, along with the release of these cells from chip for downstream analysis and/or culture. The strategy employs the use of a novel Parylene C membrane slot pore microfilter to capture CTC and a coating of poly (N-iso-propylacrylamide) (PIPAAm) for thermoresponsive viable release of the captured CTC. The capture of live cells is enabled by leveraging the design of a slot pore geometry with specific dimensions to reduce the shear stress typically associated with the filtration process. While the microfilter exhibits a high capture efficiency, the release of these cells is non-trivial. Typically, only a small percentage of cells are released when techniques such as reverse flow or cell scraping are used. The strong adhesion of these epithelial cancer cells to the Parylene C membrane is attributable to non-specific electrostatic interaction. To counteract this effect, we employed the use of PIPAAm coating and exploited its thermal responsive interfacial properties to release the cells from the filter. Blood is first filtered at room temperature. Below 32 °C, PIPAAm is hydrophilic. Thereafter, the filter is placed in either culture media or a buffer maintained at 37 °C, which results in the PIPAAm turning hydrophobic, and subsequently releasing the electrostatically bound cells.

Collaboration


Dive into the Zheng Ao's collaboration.

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

Siyang Zheng

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yu-Chong Tai

California Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge