Network


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

Hotspot


Dive into the research topics where Jennifer H. Carlson is active.

Publication


Featured researches published by Jennifer H. Carlson.


Pigment Cell & Melanoma Research | 2011

CSPG4, a potential therapeutic target, facilitates malignant progression of melanoma

Matthew Price; Leah E. Colvin Wanshura; Jianbo Yang; Jennifer H. Carlson; Bo Xiang; Guiyuan Li; Soldano Ferrone; Arkadiusz Z. Dudek; Eva A. Turley; James B. McCarthy

Chondroitin sulfate proteoglycan 4 (CSPG4), a transmembrane proteoglycan originally identified as a highly immunogenic tumor antigen on the surface of melanoma cells, is associated with melanoma tumor formation and poor prognosis in certain melanomas and several other tumor types. The complex mechanisms by which CSPG4 affects melanoma progression have started to be defined, in particular the association with other cell surface proteins and receptor tyrosine kinases (RTKs) and its central role in modulating the function of these proteins. CSPG4 is essential to the growth of melanoma tumors through its modulation of integrin function and enhanced growth factor receptor‐regulated pathways including sustained activation of ERK 1,2. This activation of integrin, RTK, and ERK1,2 function by CSPG4 modulates numerous aspects of tumor progression. CSPG4 expression has further been correlated to resistance of melanoma to conventional chemotherapeutics. This review outlines recent advances in our understanding of CSPG4‐associated cell signaling, describing the central role it plays in melanoma tumor cell growth, motility, and survival, and explores how modifying CSPG4 function and protein–protein interactions may provide us with novel combinatorial therapies for the treatment of advanced melanoma.


Cancer Research | 2009

Melanoma Proteoglycan Modifies Gene Expression to Stimulate Tumor Cell Motility, Growth, and Epithelial-to-Mesenchymal Transition

Jianbo Yang; Matthew Price; Gui Yuan Li; Menashe Bar-Eli; Ravi Salgia; Ramasamy Jagedeeswaran; Jennifer H. Carlson; Soldano Ferrone; Eva A. Turley; James B. McCarthy

Melanoma chondroitin sulfate proteoglycan (MCSP) is a plasma membrane-associated proteoglycan that facilitates the growth, motility, and invasion of tumor cells. MCSP expression in melanoma cells enhances integrin function and constitutive activation of Erk1,2. The current studies were performed to determine the mechanism by which MCSP expression promotes tumor growth and motility. The results show that MCSP expression in radial growth phase, vertical growth phase, or metastatic cell lines causes sustained activation of Erk1,2, enhanced growth, and motility which all require the cytoplasmic domain of the MCSP core protein. MCSP expression in a radial growth phase cell line also promotes an epithelial-to-mesenchymal transition based on changes in cell morphology and the expression of several epithelial-to-mesenchymal transition markers. Finally, MCSP enhances the expression of c-Met and hepatocyte growth factor, and inhibiting c-Met expression or activation limits the increased growth and motility of multiple melanoma cell lines. The studies collectively show the importance of MCSP in promoting progression by an epigenetic mechanism and they indicate that MCSP could be targeted to delay or inhibit tumor progression in patients.


Immunobiology | 2015

Hyaluronan stimulates ex vivo B lymphocyte chemotaxis and cytokine production in a murine model of fungal allergic asthma

Sumit Ghosh; Scott A. Hoselton; Steve Wanjara; Jennifer H. Carlson; James B. McCarthy; Glenn Dorsam; Jane M. Schuh

Allergic asthma is a chronic inflammatory disease of the airways characterized by excessive eosinophilic and lymphocytic inflammation with associated changes in the extracellular matrix (ECM) resulting in airway wall remodeling. Hyaluronan (HA) is a nonsulfated glycosaminoglycan ECM component that functions as a structural cushion in its high molecular mass (HMM) but has been implicated in metastasis and other disease processes when it is degraded to smaller fragments. However, relatively little is known about the role HA in mediating inflammatory responses in allergy and asthma. In the present study, we used a murine Aspergillus fumigatus inhalational model to mimic human disease. After observing in vivo that a robust B cell recruitment followed a massive eosinophilic egress to the lumen of the allergic lung and corresponded with the detection of low molecular mass HA (LMM HA), we examined the effect of HA on B cell chemotaxis and cytokine production in the ex vivo studies. We found that LMM HA functioned through a CD44-mediated mechanism to elicit chemotaxis of B lymphocytes, while high molecular mass HA (HMM HA) had little effect. LMM HA, but not HMM HA, also elicited the production of IL-10 and TGF-β1 in these cells. Taken together, these findings demonstrate a critical role for ECM components in mediating leukocyte migration and function which are critical to the maintenance of allergic inflammatory responses.


Cancer Research | 2015

Abstract 1940: Dual inhibition of mTORC1/C2 and HER2 results in maximal antitumor efficacy in preclinical model of HER2+ breast cancer resistant to trastuzumab therapy

Pradip De; Yuliang Sun; Jennifer H. Carlson; Xiaoqian Lin; Nandini Dey; Brian Leyland-Jones

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: Approximately 30% of HER2+ breast cancers (BC) harbor activating mutations (helical or kinase domain) in PIK3CA (Ellis and Perou Cancer Discovery 2013 3:27) and PIK3CA mutation is associated with lower rates of pathologic complete response to anti-HER2 therapies in primary HER2+ BC (Loibl S et al JCO 2014 55:7876). The hypothesis is that the suppression of one of the nodal points of this pathway results in sensitization to anti-HER2 agents. However, the combination of trastuzumab (T) and mTORC1/C2 inhibitor has not been comprehensively tested in T-sensitive and HER2+/PIK3CA mutated models. Methodology: Here we investigate the preclinical efficacy of a dual mTORC1/C2 inhibitor (MLN-0128, formally called INK128) alone or in combination with T in HER2+/T-sensitive/PIK3CA wild type (neither helical nor kinase domain mutation) (BT474), and HER2+/PIK3CA mutated (HCC1954, UACC893, MDA-MB453) cell lines. We analyzed in vitro cell viability and induction of apoptosis in all HER2+ cell lines. PI3K signaling was examined by immunoblot for phosphoproteins in HER2-PI3K-AKT-mTOR signaling cascade. Tumor growth inhibition was evaluated after the treatment with T, MLN-0128 or the combination in HER2 amplified models with wild type or mutant PIK3CA. Results: Our data showed that 1) PIK3CA mutation in HER2+ cells were responsible for resistance to the HER2-specific antibody T, 2) T-resistance conferred by this activating PIK3CA mutation was overcome by blockade of mTORC1/C2 with MLN-0128, 3) inhibition of phosphorylation of AKT(S473), P70S6K, S6RP, and 4EBP1(T37/46, T70) was observed following MLN-0128 treatment and the combination of MLN-0128 and T more effectively blocked the mTORC1/C2 signaling pathway, 4) MLN-0128 induced apoptosis of both BT474 and HCC1954 cells as indicated by Annexin V staining, 5) in vivo, both cell line-based xenografts showed exquisite sensitivity to the antitumor activity of the combination of T and MLN-0128, which resulted in durable tumor shrinkage and exhibited no signs of toxicity in these models and 6) immununo-staining of tumor tissues from MLN-0128 treated mice showed growth inhibition (Ki67), tumor-induced angiogenesis (CD31) and a decreased in associated phospho-proteins (p-S6RP, p-4EBP1). Conclusion: These results suggest that the combination of MLN-0128 plus T may be a potential strategy for the treatment of T-resistant breast cancer mediated by activating mutation of PIK3CA and this combination also provides benefit to HER2+/PIK3CA wild type tumors. Citation Format: Pradip Kr De, Yuliang Sun, Jennifer H. Carlson, Xiaoqian Lin, Nandini Dey, Brian Leyland-Jones. Dual inhibition of mTORC1/C2 and HER2 results in maximal antitumor efficacy in preclinical model of HER2+ breast cancer resistant to trastuzumab therapy. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1940. doi:10.1158/1538-7445.AM2015-1940


Cancer Research | 2017

Abstract 1348: GDC-0032, a p110 beta sparing PI3K inhibitor is highly efficient on PIK3CA-mutated and HER2-amplified breast cancer model

Pradip De; Yuliang Sun; Jennifer H. Carlson; Lori S. Friedman; Casey Williams; Nandini Dey; Brian Leyland-Jones

Background: HER2 amplification/overexpression frequently co-occurs with PI3K pathway activation in breast tumors. Mutations in PIK3CA are known to be involved in a wide range of human cancers including HER2+ breast cancer, and mutant PIK3CA is thought to act as an oncogene. Here, we evaluate the efficacy of GDC0032, a p110 beta-sparing PI3K inhibitor against HER2+ or HER2+/PIK3CA mutated breast cancer cells. MATERIALS AND METHODS: Four HER2+ breast cancer (BC) cell lines consisting of three activating (either helical or kinase domain) PIK3CA mutation (HER2+/ER+ and HER2+/ER-) were analyzed for proliferation, apoptosis, cell cycle and signaling pathway activation assays. Preclinical efficacy of GDC-0032 was also evaluated in vivo in a mouse model. RESULTS: 1) All HER2+ and HER2+/PIK3CA mutant cell lines exhibited low IC50 values (ranging from 0.1 µM-1.5 µM) irrespective of ER-positive or not. 2) GDC-0032 caused a strong differential growth inhibition in both HER2+ and HER2+/ /PIK3CA mutated breast cancer cell lines when compared to lines that were HER2- and PIK3CA wild type by 3D-ON-TOP clonogenic assay. 3) Administration of GDC-0032 induced cell cycle G0/G1 arrest and resulted in increased apoptosis in a dose-dependent manner. Furthermore, induction of apoptosis was more with GDC-0032 when combined with RAD001. 4) GDC0032 also blocked expression of CYCLIN D1. 5) Investigation of the signal transduction revealed that the treatment of GDC-0032 reduced the level of p-AKT (Ser473 and Thr308), and p-S6 expression is indicating the down-regulation of downstream signaling of PI3K and mTOR pathway. 6) GDC-0032 was highly active at reducing established tumor growth in vivo in BC mouse xenografts harboring PIK3CA mutation and HER2 amplification. CONCLUSION: Our in vitro or in vivo data showed that GDC-0032 was highly efficient either as a single agent as well as with RAD001 in HER2+/PIK3CA mutated breast cancer cell lines. Our data suggest that GDC-0032 represents a novel therapeutic option in patients harboring PIK3CA mutations and/or HER2/neu gene amplification in breast cancer. Citation Format: Pradip De, Yuliang Sun, Jennifer Carlson, Lori Friedman, Casey Williams, Nandini Dey, Brian Leyland-Jones. GDC-0032, a p110 beta sparing PI3K inhibitor is highly efficient on PIK3CA-mutated and HER2-amplified breast cancer model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 1348. doi:10.1158/1538-7445.AM2017-1348


Archive | 2016

PI3K-AKT-mTOR Pathway Cooperates with the DNA Damage Repair Pathway: Carcinogenesis in Triple-Negative Breast Cancers and Beyond

Pradip De; Jennifer H. Carlson; Brian Leyland-Jones; Nandini Dey

Understanding of the biology of tumor cells of breast cancers like most of other solid tumors has undergone an evolutionary change at the molecular level with the advent of exome sequencing or whole genome sequencing technology. In today’s “clinic to laboratory and back” culture of medical practice, the genomics alteration-driven (biomarker) approach to cancer treatment has not only provided a better justification for the treatment of breast cancers but also added an invaluable tool for the diagnosis of “cancer evolution” and its therapeutic management. Recognizing the fact that the PI3K-mTOR pathway is a master regulator of tumor cell survival, proliferation, nutrient sensor, protein translation, metabolism, metastasis-associated phenotypes and angiogenesis, the PI3K pathway has been explored as one of the most studied targets in cancers. We here evaluated merits of the combination of DNA damage repair pathway inhibitor(s) with inhibitor(s) of the PI3K-mTOR pathway in targeting basal-like and triple-negative breast cancers. Sensitizing triple-negative breast cancer cells to inhibitors of DNA damage repair pathway by inhibitors of PI3K-mTOR pathway has been comprehensively discussed in the light of recent studies demonstrating that PI3K-AKT-mTOR pathway closely cooperates with the DNA damage repair pathway.


Molecular Cancer Research | 2016

Abstract B12: Co-targeting PTEN-defined TNBC with p110beta-isoform specific inhibitor and PARP inhibitor BMN 673: A predictive context to sensitize PARP inhibitors in TNBC

Nandini Dey; Jennifer H. Carlson; Pradip De; Brian Leyland-Jones

Background: TNBC is a highly aggressive form of BRCA-associated BC subtype for which chemotherapy still remains a major form of treatment to which patients initially respond yet a majority of them inevitably relapse. PARP1 has been identified as a target in BRCA-defined cancers and inhibitors of PARP1 has been recently approved by FDA as targeted agents in BRCA-defined cancers. The loss of PTEN is the most common “first event” associated with basal-like subtype (Martins et al., 2012) and this PI3K-pathway activating event (deletion/mutation/loss of PTEN) occurs more frequently (35%) than PIK3CA mutations in this subtype of BC (Ellis & Perou, 2013). In the cytoplasm, PTEN-loss mediated upregulation of PI3K/AKT signal is known to depend on the PI3Kbeta-isoform. In contrast nuclear PTEN controls DNA repair (Bassi et al., 2013). A failure to repair the damaged DNA upon inhibition of PARP causes accumulation of DNA double-strand breaks and leads to apoptosis of cancer cells. Thus we hypothesized that a combination of p110beta-isoform specific inhibitor and PARP inhibitor will sensitize PARP inhibitors in PTEN-null TNBC model. Methods: Five PARP inhibitors, Talazoparib (BMN673, B), Niraparib (N), Olaparib (O), Rucaparib (R) and Veliparib (V) were tested in four PTEN-null TNBC cell lines, MDA-MB468, HCC70 (p.F90fs*9), BT549 (p.V275fs*1) and SUM149 cell lines in combination with p110beta specific inhibitor, AZD6482. Proliferative, apoptotic and PARylation signals following drug combinations were demonstrated by WB in a dose and time dependent manner. Pro-apoptotic and anti-proliferative effects were verified using complementary 3D ON-TOP assay, real time proliferation (Incucyte), AnnexinV and cl-caspase3 analyses. In order to understand the initial effects of isoform-specific inhibitors of PI3K we also compared anti-proliferative signals of GDC-0032 (p110 beta sparing inhibitor) and GDC-0941 (pan PI3K inhibitor) with that of AZD6482 at 3/6 hours in MDA-MB468 and SUM149 cells. Results: In PTEN null TNBC cells 500 nM BMN673 alone was effective in slowing cell proliferation and inducing apoptosis while AZD6482 (both 5-10 uM) did not have anti-proliferative or pro-apoptotic effects at 48 and 72 hours. Combinations of different PARPi (100nM of B, 1uM of N, 10uM of O, 10uM of R and 10uM of V) with AZD6482 blocked 3D growth of PTEN null BT549 and MDA-MB468 TNBT cells in a time dependent manner (96 hours & 7 days). PARP inhibitors abrogated PAR signals either alone or in combination with carboplatin in both BRCA1/2 WT and mutated cells. The most pronounced anti-tumor effect was observed with the combination of B and AZD6482 which was mechanistically explained by the robust increase of AnnexinV positive cells following a single 500nM dose of B at both 48 and 72 hours. In both MDA-MB468 and SUM149 cells although panPI3K inhibitor, GDC-0941 downregulated pAKT (S473, T308), pP70S6K (T389), pS6RP and p4EBP1 (T37/46) levels, p110 alpha-specific inhibitor GDC-0032 failed to perturb these signals while AZD6482 blocked them all. In summary, we demonstrate a remarkable sensitivity of tumor cells to PARP inhibition in PTEN-defined TNBC models and identify that PTEN-nullness as a potential predictive context for a possible co-targeting of PI3K pathway to further sensitize TNBC to PARP inhibitors. Significance: Our findings squarely place PTEN expression at the nexus of oncogenic signals in TNBC and indicate that TNBC patients with no PTEN activity in their tumors might benefit from combined p110beta-isoform specific inhibitor and PARP inhibitor. Citation Format: Nandini Dey, Jennifer H. Carlson, Pradip De, Brian Leyland-Jones. Co-targeting PTEN-defined TNBC with p110beta-isoform specific inhibitor and PARP inhibitor BMN 673: A predictive context to sensitize PARP inhibitors in TNBC. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B12.


Molecular Cancer Research | 2016

Abstract B11: A dual PI3K/mTOR inhibitor, BEZ235 blocks tumor-induced angiogenesis: Evidence for an effect on the tumor and endothelial compartment

Pradip De; Yuliang Sun; Jennifer H. Carlson; Xiaoqian Lin; Nandini Dey; Brian Leyland-Jones

Abstracts: AACR Special Conference: Advances in Breast Cancer; October 17-20, 2015; Bellevue, WA Background: Tumor progression including HER2+ breast tumor, particularly in aggressive and malignant tumors, is associated with the angiogenesis. To investigate the effects of BEZ235 on angiogenesis, we evaluated its effects on the hypoxic stabilization or the PI3K-AKT-mTOR pathway-mediated synthesis of hypoxia-inducible factor -1α (HIF-1α) and tumor-induced angiogenesis. Methodology: Here we have studied the in vitro and in vivo effects of BEZ235 in HER2+/T-sensitive (BT474), HER2+/T-resistant (BT474HerR) and HER2+/PIK3CA (HCC1954 & UACC893) mutated models. We assessed in vitro activation status of the PI3K-AKT-mTOR signaling pathway following BEZ235 treatment in HER2+ BC cell lines. We next evaluated the impact of BEZ235 on hypoxia-induced or the PI3K-AKT-mTOR pathway ⊠mediated HIF1α stabilization/synthesis which is a master regulator of angiogenesis and also tumor-induced angiogenesis using xenograft models. Results: The results show that 1) BEZ235 inhibited downstream activation of the PI3K-AKT-mTOR signaling pathway effectors, p-AKT (Ser473, The308), p-P70S6K, p-S6RP and p-4EBP1, 2) unlike other pan-PI3Kinase inhibitor (e.g. LY294002, SF1126), BEZ235 has no effect on hypoxia-mediated stabilization of HIF1α however, heregulin-induced HIF1α synthesis which is an important angiogenic modulator in breast cancer cells, was significantly decreased following the treatment of BEZ235 in HER2+ and HER2+/PIK3CA mutated breast cancer cells, 3) interestingly, BEZ235 mediated abrogation of HIF1α synthesis is more pronounced than mTORC1 inhibition alone, 4) integrin-directed endothelial cell migration is one of the critical steps for tumor-induced angiogenesis. BEZ235 abrogates endothelial cells functions especially integrin-directed HUVEC cells migration on vitronectin (avb3/ avb5) and significantly abrogates endothelial cells tube formation on Matrigel and 5) furthermore, we evaluated the effects of BEZ235 on the capacity of HER2+ and HER2+/PIK3CA mutated breast tumor cells to recruit a blood supply in vivo during thrice weekly treatments with BEZ235. A microvessel density (MVD) in control versus BEZ235-treated tumors showed a significant decrease in MVD (CD31+) and also VEGFR immunostaining in BEZ235 treated tumors. Conclusion: Our data suggest that BEZ235 has potent antiangiogenic activity in three different xenograft models probably via mTOR-HIF1α-VEGF signaling axis. Citation Format: Pradip De, Yuliang Sun, Jennifer H. Carlson, Xiaoqian Lin, Nandini Dey, Brian Leyland-Jones. A dual PI3K/mTOR inhibitor, BEZ235 blocks tumor-induced angiogenesis: Evidence for an effect on the tumor and endothelial compartment. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B11.


Cancer Research | 2016

Abstract 3303: Identifying a functional correlation between cancer stem cells and 3D clonogenic growth: testing rational combinations of PI3K pathway inhibitors in subtype-specific BC

Jennifer H. Carlson; Nandini Dey; Pradip De; Lori Friedman; Casey Williams; Brian Leyland-Jones

Introduction: Studying clonogenic behavior of tumor cells in the context of physiologically relevant ECM offers the potential to explore signaling processes which more closely approximate the in vivo microenvironment. This approach is valuable for identification and dissection of the key molecular signaling of cancer cells to test targeted drugs. 3D cell cultures have been recognized as the method of choice for the physiologically relevant modeling of many aspects of malignant cell behavior ex vivo. Kenny et al., 2007 identified that morphologies of breast cancer cell lines in 3D assays correlate with their gene expression profiles and protein expression patterns reflecting the underlying distinct morphologies which were associated with tumor cell invasiveness and metastases-associated phenotypes. Study of the molecular characteristics and clinical significance of CSC in the context of prognostic relevance as well as the understanding the scope of therapeutically targeting CSC within distinct tumor cell populations have revealed that CD44+ cell-specific genes represents SC markers and correlated with decreased patient survival (Fillmore and Kuperwasser, 2008). Hypothesis: Since 3D clonogenic growth of epithelial tumor cells represents the most appropriate model in vitro, we hypothesized that there is a functional association between phenotypic 3D clonogenic growth and cancer stem cells (CSC) proportion. Methods: To study the role of CSCs in the 3D clonogenic growth of tumor cells we have used BC and ovarian cell lines.The cells were challenged to form 3D colonies in matrigel and cells were recovered from the 3D colonies after de-gelling using PBS-EDTA buffer. Relative distribution of CD24+, CD44+, CD24- and CD44- expression in cells were determined by flow cytometry (Accuri C6) following recovery of the cells from the colony and compared to cells grown in 2D. We have tested the effect of a rational combination of pan-PI3K pathway inhibitor, GDC-0941 or isoform-specific inhibitors along with AI, trastuzumab, or HRD inhibitors (PARP) in ER+/PIK3CA mutated, HER2+/PIK3CA mutated or PTEN-null TNBC cells. Results: CD44/CD24 expression level changes between 2D and 3D growth was characterized in individual cell lines. Overall 3D growth led to a reduction in CD44 levels. Rational drug combinations blocked proliferative signals and enhanced apoptosis (cleaved caspase3) as demonstrated by WB, AnnexinV staining, proliferation (Incucyte). The mechanistic details of the effect of this combination on the CD44H/CD24L stem cell populations are being worked out, the results of which will be presented in the meeting. Significance: Our study reports a methodological breakthrough to determine CD24L/CD44H CSC fraction from live cells following 3D clonogenic transformation. Citation Format: Jennifer H. Carlson, Nandini Dey, Pradip De, Lori Friedman, Casey Williams, Brian Leyland-Jones. Identifying a functional correlation between cancer stem cells and 3D clonogenic growth: testing rational combinations of PI3K pathway inhibitors in subtype-specific BC. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 3303.


Cancer Research | 2016

Abstract 4605: Metastasis-associated tumor cell phenotypes in TNBC is regulated by Wnt-beta-catenin pathway signals

Nandini Dey; Jennifer H. Carlson; Pradip De; Brian Leyland-Jones

Introduction: Acquisitions of metastasis-associated phenotypes (MA) by Triple Negative Breast Cancer (TNBC) cells are imparted by genetic alterations which cause deregulation of different signaling pathways associated to those phenotypes. We have recently reported that an upregulation of the Wnt-beta-catenin pathway (WP) is one of the salient genetic features of TNBC and demonstrated that the WP signaling in TNBC is associated with poor prognosis and metastasis (Dey et al., 2013). We have also identified that the functional upregulation of secreted-MMP7, a transcriptional target of WP in TNBC is associated to the functional loss/absence of PTEN gene (Dey et al., 2013), the most common first event associated with basal-like subtype (Martins et al., 2012). Aim: In order to understand the relevance of WP pathway in the biology of metastasizing TNBC tumor cells, here we have undertaken a study in which the involvement of WP is tested in the context of MA phenotypes including (1) integrin-directed migration, (2) matrigel-invasion and (3) clonogenic growth and proliferation in 15 TNBC cell lines. Methods: CHIR9902, Wnt-C59, XAV939, sulindac sulfide, beta-catenin SiRNA as well as physiological stimulation of the WP (LWnt3A CM) were used for the study. Podia-parameters were studied using confocal microscopy for architecture of filamentous actin while matrigel invasion was studied by MMP7 specific caesin-zymography. Results: The collective% of alterations in CTNNB1, APC and DVL1 genes among total breast invasive carcinomas (TCGA 2012) were 17% in contrast to 46% breast invasive carcinomas, PAM50 Basal. A similar trend was observed among subtypes of BC from brca/tcga/pub2015 (cBioPortal). Wnt-C59, XAV939, sulindac sulfide and beta-catenin SiRNA (1) inhibited fibronectin-directed migration (2) decreased maximum relative distance from the origin and average velocity of the movement by real-time video microscopy, (3) altered cytoskeletal organization of the filamentous-actin and decreased the podia parameters, (4) decreased matrigel-invasion, and (6) inhibited cell proliferation and 3D colony formation. Mechanistically the treatment with sulindac sulfide and beta-catenin SiRNA decreased cellular levels of beta-catenin, active beta-catenin and MMP7 in the same TNBC cell lines wherein fibronectin-directed migration, invasion, and colony formation were found inhibited. LWnt3ACM stimulated cell proliferation, clonogenicity, migration and invasion was perturbed by WP modulators, sulindac sulfide and PI3K pathway inhibitor in brain-metastasis specific TNBC cells. Conclusion: Here we present the first conclusive evidence to demonstrate that the WP activation is functionally responsible for MA phenotypes in TNBC. Our data explains the why different components of the WP is upregulated in TNBC and how WP activation is associated with poor prognosis and metastasis in this subtype. Citation Format: Nandini Dey, Jennifer H. Carlson, Pradip De, Brian R. Leyland-Jones. Metastasis-associated tumor cell phenotypes in TNBC is regulated by Wnt-beta-catenin pathway signals. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4605.

Collaboration


Dive into the Jennifer H. Carlson'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

Jianbo Yang

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Lori Friedman

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge