Network


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

Hotspot


Dive into the research topics where Amber Ablack is active.

Publication


Featured researches published by Amber Ablack.


Cell Reports | 2014

Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis.

Hon S. Leong; Amy E. Robertson; Konstantin Stoletov; Sean J. Leith; Curtis A. Chin; Andrew E. Chien; M. Nicole Hague; Amber Ablack; Katia Carmine-Simmen; Victor A. McPherson; Carl O. Postenka; Eva A. Turley; Sara A. Courtneidge; Ann F. Chambers; John D. Lewis

Tumor cell extravasation is a key step during cancer metastasis, yet the precise mechanisms that regulate this dynamic process are unclear. We utilized a high-resolution time-lapse intravital imaging approach to visualize the dynamics of cancer cell extravasation in vivo. During intravascular migration, cancer cells form protrusive structures identified as invadopodia by their enrichment of MT1-MMP, cortactin, Tks4, and importantly Tks5, which localizes exclusively to invadopodia. Cancer cells extend invadopodia through the endothelium into the extravascular stroma prior to their extravasation at endothelial junctions. Genetic or pharmacological inhibition of invadopodia initiation (cortactin), maturation (Tks5), or function (Tks4) resulted in an abrogation of cancer cell extravasation and metastatic colony formation in an experimental mouse lung metastasis model. This provides direct evidence of a functional role for invadopodia during cancer cell extravasation and distant metastasis and reveals an opportunity for therapeutic intervention in this clinically important process.


Small | 2011

Intravital imaging of human prostate cancer using viral nanoparticles targeted to gastrin-releasing Peptide receptors.

Nicole F. Steinmetz; Amber Ablack; Jennifer L. Hickey; Jailal N. G. Ablack; Bhavik Manocha; Joe S. Mymryk; Leonard G. Luyt; John D. Lewis

Multivalent nanoparticles have several key advantages in terms of solubility, binding avidity, and uptake, making them particularly well suited to molecular imaging applications. Herein is reported the stepwise synthesis and characterization of NIR viral nanoparticles targeted to gastrin-releasing peptide receptors that are over-expressed in human prostate cancers. The pan-bombesin analogue, [β-Ala11, Phe13, Nle14]bombesin-(7-14), is conjugated to cowpea mosaic virus particles functionalized with an NIR dye (Alexa Fluor 647) and polyethylene glycol (PEG) using the copper(I)-catalyzed azide-alkyne cycloaddition reaction. Targeting and uptake in human PC-3 prostate cells is demonstrated in vitro. Tumor homing is observed using human prostate tumor xenografts on the chicken chorioallantoic membrane model using intravital imaging. Further development of this viral nanoparticle platform may open the door to potential clinical noninvasive molecular imaging strategies.


Nature Protocols | 2010

Intravital imaging of embryonic and tumor neovasculature using viral nanoparticles

Hon S. Leong; Nicole F. Steinmetz; Amber Ablack; Giuseppe Destito; Andries Zijlstra; Heidi Stuhlmann; Marianne Manchester; John D. Lewis

Viral nanoparticles are a novel class of biomolecular agents that take advantage of the natural circulatory and targeting properties of viruses to allow the development of therapeutics, vaccines and imaging tools. We have developed a multivalent nanoparticle platform based on the cowpea mosaic virus (CPMV) that facilitates particle labeling at high density with fluorescent dyes and other functional groups. Compared with other technologies, CPMV-based viral nanoparticles are particularly suited for long-term intravital vascular imaging because of their biocompatibility and retention in the endothelium with minimal side effects. The stable, long-term labeling of the endothelium allows the identification of vasculature undergoing active remodeling in real time. In this study, we describe the synthesis, purification and fluorescent labeling of CPMV nanoparticles, along with their use for imaging of vascular structure and for intravital vascular mapping in developmental and tumor angiogenesis models. Dye-labeled viral nanoparticles can be synthesized and purified in a single day, and imaging studies can be conducted over hours, days or weeks, depending on the application.


Molecular Pharmaceutics | 2013

Increased Tumor Homing and Tissue Penetration of the Filamentous Plant Viral Nanoparticle Potato virus X

Sourabh Shukla; Amber Ablack; Amy M. Wen; Karin L. Lee; John D. Lewis; Nicole F. Steinmetz

Nanomaterials with elongated architectures have been shown to possess differential tumor homing properties compared to their spherical counterparts. Here, we investigate whether this phenomenon is mirrored by plant viral nanoparticles that are filamentous (Potato virus X) or spherical (Cowpea mosaic virus). Our studies demonstrate that Potato virus X (PVX) and Cowpea mosaic virus (CPMV) show distinct biodistribution profiles and differ in their tumor homing and penetration efficiency. Analogous to what is seen with inorganic nanomaterials, PVX shows enhanced tumor homing and tissue penetration. Human tumor xenografts exhibit higher uptake of PEGylated filamentous PVX compared to CPMV, particularly in the core of the tumor. This is supported by immunohistochemical analysis of the tumor sections, which indicates greater penetration and accumulation of PVX within the tumor tissues. The enhanced tumor homing and retention properties of PVX along with its higher payload carrying capacity make it a potentially superior platform for applications in cancer drug delivery and imaging applications.


Nanomedicine: Nanotechnology, Biology and Medicine | 2011

Cowpea mosaic virus nanoparticles target surface vimentin on cancer cells

Nicole F. Steinmetz; Choi-Fong Cho; Amber Ablack; John D. Lewis; Marianne Manchester

AIMS Vimentin, a type III intermediate filament, is upregulated during epithelial-mesenchymal transition and tumor progression. Vimentin is surface-expressed on cells involved in inflammation; the function remains unknown. We investigated the expression of surface vimentin on cancer cells and evaluated targeting nanoparticles to tumors exploiting vimentin. MATERIALS & METHODS Cowpea mosaic virus nanoparticles that interact with surface vimentin were used as probes. Tumor homing was tested using the chick chorioallantoic membrane model with human tumor xenografts. RESULTS & DISCUSSION Surface vimentin levels varied during cell cycle and among the cell lines tested. Surface vimentin expression correlated with cowpea mosaic virus uptake, underscoring the utility of cowpea mosaic virus to detect invasive cancer cells. Targeting to tumor xenografts was observed; homing was based on the enhanced permeability and retention effect. Our data provide novel insights into the role of surface vimentin in cancer and targeting nanoparticles in vivo.


Journal of Biological Chemistry | 2012

Loss of Pannexin 1 Attenuates Melanoma Progression by Reversion to a Melanocytic Phenotype

Silvia Penuela; Laszlo Gyenis; Amber Ablack; Jared M. Churko; Amy C. Berger; David W. Litchfield; John D. Lewis; Dale W. Laird

Background: Panx1 is a channel-forming glycoprotein that regulates epidermal differentiation and proliferation. Results: Depletion of Panx1 in melanomas causes cell re-differentiation into a melanocytic-like phenotype and reduced tumorigenesis. Conclusion: Panx1 is up-regulated during melanoma progression promoting tumor growth and metastasis. Significance: This is the first report of Panx1 as a proto-oncogene establishing it as a potential target for melanoma treatment. Pannexin 1 (Panx1) is a channel-forming glycoprotein expressed in different cell types of mammalian skin. We examined the role of Panx1 in melanoma tumorigenesis and metastasis since qPCR and Western blots revealed that mouse melanocytes exhibited low levels of Panx1 while increased Panx1 expression was correlated with tumor cell aggressiveness in the isogenic melanoma cell lines (B16-F0, -F10, and -BL6). Panx1 shRNA knockdown (Panx1-KD) generated stable BL6 cell lines, with reduced dye uptake, that showed a marked increase in melanocyte-like cell characteristics including higher melanin production, decreased cell migration and enhanced formation of cellular projections. Western blotting and proteomic analyses using 2D-gel/mass spectroscopy identified vimentin and β-catenin as two of the markers of malignant melanoma that were down-regulated in Panx1-KD cells. Xenograft Panx1-KD cells grown within the chorioallantoic membrane of avian embryos developed tumors that were significantly smaller than controls. Mouse-Alu qPCR of the excised avian embryonic organs revealed that tumor metastasis to the liver was significantly reduced upon Panx1 knockdown. These data suggest that while Panx1 is present in skin melanocytes it is up-regulated during melanoma tumor progression, and tumorigenesis can be inhibited by the knockdown of Panx1 raising the possibility that Panx1 may be a viable target for the treatment of melanoma.


Laboratory Investigation | 2011

Nuclear localization of maspin is essential for its inhibition of tumor growth and metastasis

Brigitte Goulet; Wendy Kennette; Amber Ablack; Carl O. Postenka; M. Nicole Hague; Joe S. Mymryk; Alan B. Tuck; Vincent Giguère; Ann F. Chambers; John D. Lewis

Maspin (mammary serine protease inhibitor or SerpinB5) acts as a tumor suppressor when overexpressed in aggressive cancer cell lines. However, its role in human cancer is controversial. Maspin expression has been associated with a poor prognosis in some studies, whereas in others, with favorable outcome. The clinical data suggest, however, that nuclear-localized maspin is associated with improved survival. We hypothesized that the tumor suppressor activity of maspin may require nuclear localization, and that the discordance between clinical and experimental reports is a consequence of the variable subcellular distribution of maspin. Furthermore, we surmized that nuclear maspin could function as a tumor suppressor through the regulation of genes involved in tumor growth and invasion. Maspin or maspin fused to a nuclear export signal were expressed in metastatic human breast and epidermoid carcinoma cell lines. We found that pan-cellular localized maspin inhibited in vivo tumor growth and metastasis when assessed in xenograft chicken embryo and murine mammary fat pad injection models. However, when maspin was excluded from the nucleus via a nuclear exclusion signal, it no longer functioned as a metastasis suppressor. Using chromatin immunoprecipitation, we show that nuclear maspin was enriched at the promoter of colony-stimulating factor-1 (CSF-1) and associated with diminished levels of CSF-1 mRNA. Our findings demonstrate that the nuclear localization of maspin is required for its tumor and metastasis suppressor functions in vivo, and suggest that its mechanism of action involves, in part, direct association of maspin with target genes.


Cancer Research | 2012

Embryonic Protein Nodal Promotes Breast Cancer Vascularization

Daniela F. Quail; Logan A. Walsh; Guihua Zhang; Scott D. Findlay; Juan Moreno; Laura Fung; Amber Ablack; John D. Lewis; Susan J. Done; David A. Hess; Lynne-Marie Postovit

Tumor vascularization is requisite for breast cancer progression, and high microvascular density in tumors is a poor prognostic indicator. Patients bearing breast cancers expressing human embryonic stem cell (hESC)-associated genes similarly exhibit high mortality rates, and the expression of embryonic proteins is associated with tumor progression. Here, we show that Nodal, a hESC-associated protein, promotes breast cancer vascularization. We show that high levels of Nodal are positively correlated with high vascular densities in human breast lesions (P = 0.0078). In vitro, we show that Nodal facilitates breast cancer-induced endothelial cell migration and tube formation, largely by upregulating the expression and secretion of proangiogenic factors by breast cancer cells. Using a directed in vivo angiogenesis assay and a chick chorioallantoic membrane assay, we show that Nodal promotes vascular recruitment in vivo. In a clinically relevant in vivo model, whereby Nodal expression was inhibited following tumor formation, we found a significant reduction in tumor vascularization concomitant with elevated hypoxia and tumor necrosis. These findings establish Nodal as a potential target for the treatment of breast cancer angiogenesis and progression.


Journal of Visualized Experiments | 2011

Evaluation of Nanoparticle Uptake in Tumors in Real Time Using Intravital Imaging

Choi-Fong Cho; Amber Ablack; Hon-Sing Leong; Andries Zijlstra; John D. Lewis

Current technologies for tumor imaging, such as ultrasound, MRI, PET and CT, are unable to yield high-resolution images for the assessment of nanoparticle uptake in tumors at the microscopic level1,2,3, highlighting the utility of a suitable xenograft model in which to perform detailed uptake analyses. Here, we use high-resolution intravital imaging to evaluate nanoparticle uptake in human tumor xenografts in a modified, shell-less chicken embryo model. The chicken embryo model is particularly well-suited for these in vivo analyses because it supports the growth of human tumors, is relatively inexpensive and does not require anesthetization or surgery 4,5. Tumor cells form fully vascularized xenografts within 7 days when implanted into the chorioallantoic membrane (CAM) 6. The resulting tumors are visualized by non-invasive real-time, high-resolution imaging that can be maintained for up to 72 hours with little impact on either the host or tumor systems. Nanoparticles with a wide range of sizes and formulations administered distal to the tumor can be visualized and quantified as they flow through the bloodstream, extravasate from leaky tumor vasculature, and accumulate at the tumor site. We describe here the analysis of nanoparticles derived from Cowpea mosaic virus (CPMV) decorated with near-infrared fluorescent dyes and/or polyethylene glycol polymers (PEG) 7, 8, 9,10,11. Upon intravenous administration, these viral nanoparticles are rapidly internalized by endothelial cells, resulting in global labeling of the vasculature both outside and within the tumor7,12. PEGylation of the viral nanoparticles increases their plasma half-life, extends their time in the circulation, and ultimately enhances their accumulation in tumors via the enhanced permeability and retention (EPR) effect 7, 10,11. The rate and extent of accumulation of nanoparticles in a tumor is measured over time using image analysis software. This technique provides a method to both visualize and quantify nanoparticle dynamics in human tumors.


PLOS ONE | 2012

Imaging the Impact of Chemically Inducible Proteins on Cellular Dynamics In Vivo

Hon S. Leong; Michael M. Lizardo; Amber Ablack; Victor A. McPherson; Thomas J. Wandless; Ann F. Chambers; John D. Lewis

The analysis of dynamic events in the tumor microenvironment during cancer progression is limited by the complexity of current in vivo imaging models. This is coupled with an inability to rapidly modulate and visualize protein activity in real time and to understand the consequence of these perturbations in vivo. We developed an intravital imaging approach that allows the rapid induction and subsequent depletion of target protein levels within human cancer xenografts while assessing the impact on cell behavior and morphology in real time. A conditionally stabilized fluorescent E-cadherin chimera was expressed in metastatic breast cancer cells, and the impact of E-cadherin induction and depletion was visualized using real-time confocal microscopy in a xenograft avian embryo model. We demonstrate the assessment of protein localization, cell morphology and migration in cells undergoing epithelial-mesenchymal and mesenchymal-epithelial transitions in breast tumors. This technique allows for precise control over protein activity in vivo while permitting the temporal analysis of dynamic biophysical parameters.

Collaboration


Dive into the Amber Ablack's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicole F. Steinmetz

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Ann F. Chambers

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Hon S. Leong

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jailal N. G. Ablack

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Jennifer L. Hickey

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Joe S. Mymryk

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Leonard G. Luyt

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Amy C. Berger

University of Western Ontario

View shared research outputs
Top Co-Authors

Avatar

Dale W. Laird

University of Western Ontario

View shared research outputs
Researchain Logo
Decentralizing Knowledge