Network


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

Hotspot


Dive into the research topics where Konstantin Stoletov is active.

Publication


Featured researches published by Konstantin Stoletov.


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.


Journal of Biological Chemistry | 2014

Protein-tyrosine Pseudokinase 7 (PTK7) Directs Cancer Cell Motility and Metastasis

Vladislav S. Golubkov; Natalie L. Prigozhina; Yong Zhang; Konstantin Stoletov; John D. Lewis; Phillip E. Schwartz; Robert M. Hoffman; Alex Y. Strongin

Background: In embryonic development, PTK7 regulates orientation of cells in a tissue plane. Results: PTK7 controls cellular protrusions and, as a result, directional cell motility and metastasis in fibrosarcoma HT1080 cells. Conclusion: Both PTK7 expression and proteolysis contribute to efficient cell motility and metastasis. Significance: PTK7 is a potential diagnostic biomarker with predictive value and a promising drug target in cancer. It is well established that widely expressed PTK7 is essential for vertebrate tissue morphogenesis. In cancer, the functionality of PTK7 is selectively regulated by membrane type-1 matrix metalloproteinase (MT1-MMP), ADAMs (a disintegrin domain and metalloproteinases), and γ-secretase proteolysis. Here, we established that the full-length membrane PTK7, its Chuzhoi mutant with the two functional MT1-MMP cleavage sites, and its L622D mutant with the single inactivated MT1-MMP cleavage site differentially regulate cell motility in a two-dimensional versus three-dimensional environment. We also demonstrated that in polarized cancer cells, the levels of PTK7 expression and proteolysis were directly linked to the structure and kinetics of cell protrusions, including lamellipodia and invadopodia. In the functionally relevant and widely accepted animal models of metastasis, mouse and chick embryo models, both the overexpression and knock-out of PTK7 in HT1080 cells abrogated metastatic dissemination. Our analysis of human tissue specimens confirmed intensive proteolysis of PTK7 in colorectal cancer tumors, but not in matching normal tissue. Our results provide convincing evidence that both PTK7 expression and proteolysis, rather than the level of the cellular full-length PTK7 alone, contribute to efficient directional cell motility and metastasis in cancer.


Expert Review of Anticancer Therapy | 2015

Invadopodia: a new therapeutic target to block cancer metastasis

Konstantin Stoletov; John D. Lewis

Cancer cells become dangerous when they acquire the ability to invade through physical barriers in the body and disseminate to distant sites. Recent evidence has demonstrated that cancer cells utilize specialized structures called invadopodia, unique protrusions that concentrate proteases such as matrix metalloproteinases (MMPs), to escape blood vessels during the process of extravasation. Perhaps most exciting is the fact that inhibition of invadopodia through genetic or pharmacological means reduces the ability of cancer cells to extravasate and effectively blocks metastasis. This opens the door for the development of novel therapies targeting invadopodia and cancer metastasis.


Oncotarget | 2017

Functional assessment of von Willebrand factor expression by cancer cells of non-endothelial origin

Anahita Mojiri; Konstantin Stoletov; Maria Areli Lorenzana Carrillo; Lian Willetts; Saket Jain; Roseline Godbout; Paul Jurasz; Consolato Sergi; David D. Eisenstat; John D. Lewis; Nadia Jahroudi

Von Willebrand factor (VWF) is a highly adhesive procoagulant molecule that mediates platelet adhesion to endothelial and subendothelial surfaces. Normally it is expressed exclusively in endothelial cells (ECs) and megakaryocytes. However, a few studies have reported VWF detection in cancer cells of non-endothelial origin, including osteosarcoma. A role for VWF in cancer metastasis has long been postulated but evidence supporting both pro- and anti-metastatic roles for VWF has been presented. We hypothesized that the role of VWF in cancer metastasis is influenced by its cellular origin and that cancer cell acquisition of VWF expression may contribute to enhanced metastatic potential. We demonstrated de novo expression of VWF in glioma as well as osteosarcoma cells. Endothelial monolayer adhesion, transmigration and extravasation capacities of VWF expressing cancer cells were shown to be enhanced compared to non-VWF expressing cells, and were significantly reduced as a result of VWF knock down. VWF expressing cancer cells were also detected in patient tumor samples of varying histologies. Analyses of the mechanism of transcriptional activation of the VWF in cancer cells demonstrated a pattern of trans-activating factor binding and epigenetic modifications consistent overall with that observed in ECs. These results demonstrate that cancer cells of non-endothelial origin can acquire de novo expression of VWF, which can enhance processes, including endothelial and platelet adhesion and extravasation, that contribute to cancer metastasis.


Nature Communications | 2017

LPP is a Src substrate required for invadopodia formation and efficient breast cancer lung metastasis

Elaine Ngan; Konstantin Stoletov; Harvey W. Smith; Jessica Common; William J. Muller; John D. Lewis; Peter M. Siegel

We have previously shown that lipoma preferred partner (LPP) mediates TGFβ-induced breast cancer cell migration and invasion. Herein, we demonstrate that diminished LPP expression reduces circulating tumour cell numbers, impairs cancer cell extravasation and diminishes lung metastasis. LPP localizes to invadopodia, along with Tks5/actin, at sites of matrix degradation and at the tips of extravasating breast cancer cells as revealed by intravital imaging of the chick chorioallantoic membrane (CAM). Invadopodia formation, breast cancer cell extravasation and metastasis require an intact LPP LIM domain and the ability of LPP to interact with α-actinin. Finally, we show that Src-mediated LPP phosphorylation at specific tyrosine residues (Y245/301/302) is critical for invadopodia formation, breast cancer cell invasion and metastasis. Together, these data define a previously unknown function for LPP in the formation of invadopodia and reveal a requirement for LPP in mediating the metastatic ability of breast cancer cells.


Journal of Molecular Medicine | 2016

Ankyrin G expression is associated with androgen receptor stability, invasiveness, and lethal outcome in prostate cancer patients

Tingting Wang; Hatem Abou-Ouf; Samar A. Hegazy; Mohammed Alshalalfa; Konstantin Stoletov; John D. Lewis; Bryan Donnelly; Tarek A. Bismar

Ankyrin G (ANK3) is a member of the Ankyrin family, which functions to provide cellular stability by anchoring the cytoskeleton to the plasma membrane. Deregulation of ANK3 expression has been observed in multiple human cancers but its mechanism remains unknown. ANK3 expression in relation to disease progression and patients’ outcome was investigated in two cohorts of prostate cancer (PCA). Mechanistic studies were carried out in vitro and in vivo using several PCA cell lines and the avian embryo model. Silencing ANK3 resulted in significant reduction of cell proliferation through an AR-independent mechanism. Decreased ANK3 expression delayed S phase to G2/M cell cycle transition and reduced the expression of cyclins A and B. However, cells with knocked-down ANK3 exhibited significant increase in cell invasion through an AR-dependent mechanism. Furthermore, we found that ANK3 is a regulator of AR protein stability. ANK3 knockdown also promoted cancer cell invasion and extravasations in vivo using the avian embryo model (pxa0<xa00.01). In human samples, ANK3 expression was dramatically upregulated in high grade intraepithelial neoplasia (HGPIN) and localized PCA (pxa0<xa00.0001). However, it was downregulated castration resistant stage (pxa0<xa00.0001) and showed inverse relation to Gleason score (pxa0<xa00.0001). In addition, increased expression of ANK3 in cancer tissues was correlated with better cancer-specific survival of PCA patients (pxa0=xa00.012).Key messageSilencing ANK3 results in significant reduction of cell proliferation through an AR-independent mechanism.ANK3 knockdown results in significant increase in cell invasion through an AR-dependent mechanism.ANK3 is a regulator of AR protein stability.ANK3 knockdown also promotes cancer cell invasion and extravasation in vivo using the avian embryo model.


Nature Communications | 2018

Quantitative in vivo whole genome motility screen reveals novel therapeutic targets to block cancer metastasis

Konstantin Stoletov; Lian Willetts; Robert J. Paproski; David Bond; Srijan Raha; Juan Jovel; Benjamin Adam; Amy E. Robertson; Francis Wong; Emma Woolner; Deborah Sosnowski; Tarek A. Bismar; Gane Ka-Shu Wong; Andries Zijlstra; John D. Lewis

Metastasis is the most lethal aspect of cancer, yet current therapeutic strategies do not target its key rate-limiting steps. We have previously shown that the entry of cancer cells into the blood stream, or intravasation, is highly dependent upon in vivo cancer cell motility, making it an attractive therapeutic target. To systemically identify genes required for tumor cell motility in an in vivo tumor microenvironment, we established a novel quantitative in vivo screening platform based on intravital imaging of human cancer metastasis in ex ovo avian embryos. Utilizing this platform to screen a genome-wide shRNA library, we identified a panel of novel genes whose function is required for productive cancer cell motility in vivo, and whose expression is closely associated with metastatic risk in human cancers. The RNAi-mediated inhibition of these gene targets resulted in a nearly total (>99.5%) block of spontaneous cancer metastasis in vivo.Tumour metastasis is dependent on tumour cell motility. Here, the authors investigate genes required for tumour cell motility by establishing a quantitative in vivo screening platform based on intravital imaging of human cancer metastasis in ex ovo avian embryos.


Cell Stress | 2018

Intravital imaging tumor screen used to identify novel metastasis-blocking therapeutic targets

Konstantin Stoletov; Lian Willetts; Perrin H. Beatty; John D. Lewis

Cancer cell motility is a key driver of metastasis. Although the intravasation of cancer cells into the blood stream is highly dependent on their motility and metastatic dissemination is the primary cause of cancer related deaths, current therapeutic strategies do not target the genes and proteins that are essential for cell motility. A primary reason for this is because the identification of cell motility-related genes that are relevant in vivo requires the visualization of metastatic lesions forming in an appropriate in vivo model. The cancer research community has lacked an in vivo and intravital metastatic cancer model that could be imaged as motility developed, in real-time. To address this, we developed a novel quantitative in vivo screening platform based on intravital imaging in shell-less ex ovo chick embryos. We applied this imaging approach to screen a human genome-wide short hairpin RNA library (shRNA) versus the highly motile head and neck cancer cells (HEp3 cell line) introduced into the chorioallantoic membrane (CAM) of chick embryos and identified multiple novel in vivo cancer cell motility-associated genes. When the expression of several of the identified genes was inhibited in the HEp3 tumors, we observed a nearly total block of spontaneous cancer metastasis.


Methods of Molecular Biology | 2016

Quantitative Analysis of Human Cancer Cell Extravasation Using Intravital Imaging.

Lian Willetts; David Bond; Konstantin Stoletov; John D. Lewis

Metastasis, or the spread of cancer cells from a primary tumor to distant sites, is the leading cause of cancer-associated death. Metastasis is a complex multi-step process comprised of invasion, intravasation, survival in circulation, extravasation, and formation of metastatic colonies. Currently, in vitro assays are limited in their ability to investigate these intricate processes and do not faithfully reflect metastasis as it occurs in vivo. Traditional in vivo models of metastasis are limited by their ability to visualize the seemingly sporadic behavior of where and when cancer cells spread (Reymond et al., Nat Rev Cancer 13:858-870, 2013). The avian embryo model of metastasis is a powerful platform to study many of the critical steps in the metastatic cascade including the migration, extravasation, and invasion of human cancer cells in vivo (Sung et al., Nat Commun 6:7164, 2015; Leong et al., Cell Rep 8, 1558-1570, 2014; Kain et al., Dev Dyn 243:216-28, 2014; Leong et al., Nat Protoc 5:1406-17, 2010; Zijlstra et al., Cancer Cell 13:221-234, 2008; Palmer et al., J Vis Exp 51:2815, 2011). The chicken chorioallantoic membrane (CAM) is a readily accessible and well-vascularized tissue that surrounds the developing embryo. When the chicken embryo is grown in a shell-less, ex ovo environment, the nearly transparent CAM provides an ideal environment for high-resolution fluorescent microcopy approaches. In this model, the embryonic chicken vasculature and labeled cancer cells can be visualized simultaneously to investigate specific steps in the metastatic cascade including extravasation. When combined with the proper image analysis tools, the ex ovo chicken embryo model offers a cost-effective and high-throughput platform for the quantitative analysis of tumor cell metastasis in a physiologically relevant in vivo setting. Here we discuss detailed procedures to quantify cancer cell extravasation in the shell-less chicken embryo model with advanced fluorescence microscopy techniques.


Cancer Research | 2014

Abstract 4972: In vivo whole genome shRNA screen reveals novel targets to block cancer metastasis

Konstantin Stoletov; David Bond; Hon S. Leong; Emma Woolner; Srijan Raha; Amy E. Robertson; Francis Wong; Andries Zijlstra; John D. Lewis

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CAnnThe formation of invasive, rapidly growing metastatic lesions is a critical step in cancer metastasis, the cause of more than 90% of cancer deaths. Development of novel therapeutic approaches that block the invasion step of metastasis is one of the highest priorities for clinical cancer research. For this reason we completed the first genome-wide in vivo shRNA screen for genes that directly contribute to invasive metastatic lesion formation. Using state of the art intravital imaging, we identified over fifty novel regulators of invasive metastatic colony formation in vivo. Interactome analysis links these genes to key cellular processes including: transcriptional regulation of gene expression, mRNA processing and cytoskeletal remodeling. The target list was then prioritized based on clinical gene expression profiles that negatively correlated with key cancer endpoints including metastasis, cancer-specific and overall survival. Pharmacological and shRNA-mediated knockdown of the high priority targets in human cancer cell lines such as prostate cancer and melanoma specifically blocked cancer cell migration and invasion in vitro and in vivo. Moreover, shRNA-mediated knockdown of these genes blocked human cancer cell metastasis in the avian embryo and mouse preclinical models of metastasis. Finally, immunohistochemical analysis on clinical prostate cancer and melanoma tissue samples showed strong correlation with disease progression and metastasis. In summary, we have identified numerous novel genes that that are functionally involved in cancer invasion and metastasis that may serve as predictive markers for disease aggressiveness and represent exciting new pharmacological targets to block cancer invasion and metastasis.nnCitation Format: Konstantin Stoletov, David Bond, Hon Sing Leong, Emma Woolner, Srijan Raha, Amy Robertson, Francis Wong, Andries Zijlstra, John D. Lewis. In vivo whole genome shRNA screen reveals novel targets to block cancer metastasis. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4972. doi:10.1158/1538-7445.AM2014-4972

Collaboration


Dive into the Konstantin Stoletov's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy E. Robertson

London Health Sciences Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge