Network


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

Hotspot


Dive into the research topics where Jennifer R. Yoon is active.

Publication


Featured researches published by Jennifer R. Yoon.


Cancer Research | 2008

Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cells

Rebecca A. Whipple; Eric M. Balzer; Edward H. Cho; Michael A. Matrone; Jennifer R. Yoon; Stuart S. Martin

Solid tumor metastasis often involves detachment of epithelial carcinoma cells into the vasculature or lymphatics. However, most studies of cytoskeletal rearrangement in solid tumors focus on attached cells. In this study, we report for the first time that human breast tumor cells produce unique tubulin-based protrusions when detached from extracellular matrix. Tumor cell lines of high metastatic potential show significantly increased extension and frequency of microtubule protrusions, which we have termed tubulin microtentacles. Our previous studies in nontumorigenic mammary epithelial cells showed that such detachment-induced microtentacles are enriched in detyrosinated alpha-tubulin. However, amounts of detyrosinated tubulin were similar in breast tumor cell lines despite varying microtentacle levels. Because detyrosinated alpha-tubulin associates strongly with intermediate filament proteins, we examined the contribution of cytokeratin and vimentin filaments to tumor cell microtentacles. Increased microtentacle frequency and extension correlated strongly with loss of cytokeratin expression and up-regulation of vimentin, as is often observed during tumor progression. Moreover, vimentin filaments coaligned with microtentacles, whereas cytokeratin did not. Disruption of vimentin with PP1/PP2A-specific inhibitors significantly reduced microtentacles and inhibited cell reattachment to extracellular matrix. Furthermore, expression of a dominant-negative vimentin mutant disrupted endogenous vimentin filaments and significantly reduced microtentacles, providing specific genetic evidence that vimentin supports microtentacles. Our results define a novel model in which coordination of vimentin and detyrosinated microtubules provides structural support for the extensive microtentacles observed in detached tumor cells and a possible mechanism to promote successful metastatic spread.


Oncogene | 2010

Metastatic breast tumors express increased tau, which promotes microtentacle formation and the reattachment of detached breast tumor cells.

Michael A. Matrone; Rebecca A. Whipple; Keyata Thompson; Edward H. Cho; Michele I. Vitolo; Eric M. Balzer; Jennifer R. Yoon; Olga B. Ioffe; Kimberly C. Tuttle; Ming Tan; Stuart S. Martin

The cytoskeletal organization of detached and circulating tumor cells (CTCs) is currently not well defined and may provide potential targets for new therapies to limit metastatic tumor spread. In vivo, CTCs reattach in distant tissues by a mechanism that is tubulin-dependent and suppressed by polymerized actin. The cytoskeletal mechanisms that promote reattachment of CTCs match exactly with the mechanisms supporting tubulin microtentacles (McTN), which we have recently identified in detached breast tumor cells. In this study, we aimed to investigate how McTN formation is affected by the microtubule-associated protein, tau, which is expressed in a subset of chemotherapy-resistant breast cancers. We demonstrate that endogenous tau protein localizes to McTNs and is both necessary and sufficient to promote McTN extension in detached breast tumor cells. Tau-induced McTNs increase reattachment of suspended cells and retention of CTCs in lung capillaries. Analysis of patient-matched primary and metastatic tumors reveals that 52% possess tau expression in metastases and 26% display significantly increased tau expression over disease progression. Tau enrichment in metastatic tumors and the ability of tau to promote tumor cell reattachment through McTN formation support a model in which tau-induced microtubule stabilization provides a selective advantage during tumor metastasis.


Breast Cancer Research and Treatment | 2011

Local anesthetics inhibit kinesin motility and microtentacle protrusions in human epithelial and breast tumor cells

Jennifer R. Yoon; Rebecca A. Whipple; Eric M. Balzer; Edward H. Cho; Michael A. Matrone; Michelle Peckham; Stuart S. Martin

Detached breast tumor cells produce dynamic microtubule protrusions that promote reattachment of cells and are termed tubulin microtentacles (McTNs) due to their mechanistic distinctions from actin-based filopodia/invadopodia and tubulin-based cilia. McTNs are enriched with vimentin and detyrosinated α-tubulin, (Glu-tubulin). Evidence suggests that vimentin and Glu-tubulin are cross-linked by kinesin motor proteins. Using known kinesin inhibitors, Lidocaine and Tetracaine, the roles of kinesins in McTN formation and function were tested. Live-cell McTN counts, adhesion assays, immunofluorescence, and video microscopy were performed to visualize inhibitor effects on McTNs. Viability and apoptosis assays were used to confirm the non-toxicity of the inhibitors. Treatments of human non-tumorigenic mammary epithelial and breast tumor cells with Lidocaine or Tetracaine caused rapid collapse of vimentin filaments. Live-cell video microscopy demonstrated that Tetracaine reduces motility of intracellular GFP-kinesin and causes centripetal collapse of McTNs. Treatment with Tetracaine inhibited the extension of McTNs and their ability to promote tumor cell aggregation and reattachment. Lidocaine showed similar effects but to a lesser degree. Our current data support a model in which the inhibition of kinesin motor proteins by Tetracaine leads to the reductions in McTNs, and provides a novel mechanism for the ability of this anesthetic to decrease metastatic progression.


Oncogene | 2013

Loss of PTEN induces microtentacles through PI3K-independent activation of cofilin

Michele I. Vitolo; Amanda E. Boggs; Rebecca A. Whipple; Jennifer R. Yoon; Keyata Thompson; Michael A. Matrone; Edward H. Cho; Eric M. Balzer; Stuart S. Martin

Loss of PTEN tumor suppressor enhances metastatic risk in breast cancer, although the underlying mechanisms are poorly defined. We report that homozygous deletion of PTEN in mammary epithelial cells induces tubulin-based microtentacles (McTNs) that facilitate cell reattachment and homotypic aggregation. Treatment with contractility-modulating drugs showed that McTNs in PTEN−/− cells are suppressible by controlling the actin cytoskeleton. Because outward microtubule extension is counteracted by actin cortical contraction, increased activity of actin-severing proteins could release constraints on McTN formation in PTEN−/− cells. One such actin-severing protein, cofilin, is activated in detached PTEN−/− cells that could weaken the actin cortex to promote McTNs. Expression of wild-type cofilin, an activated mutant (S3A), and an inactive mutant (S3E) demonstrated that altering cofilin phosphorylation directly affects McTNs formation. Chemical inhibition of PI3K did not reduce McTNs or inactivate cofilin in PTEN−/− cells. Additionally, knock-in expression of the two most common PI3K-activating mutations observed in human cancer patients did not increase McTNs or activate cofilin. PTEN loss and PI3K activation also caused differential activation of the cofilin regulators, LIM-kinase1 (LIMK) and Slingshot-1L (SSH). Furthermore, McTNs were suppressed and cofilin was inactivated by restoration of PTEN in the PTEN−/− cells, indicating that both the elevation of McTNs and the activation of cofilin are specific results arising from PTEN loss. These data identify a novel mechanism by which PTEN loss could remodel the cortical actin network to facilitate McTNs that promote tumor cell reattachment and aggregation. Using isogenic MCF-10A PTEN−/− and PIK3CA mutants, we have further demonstrated that there are clear differences in activation of cofilin, LIMK and SSH between PTEN loss and PI3K activation, providing a new evidence that these mutations yield distinct cytoskeletal phenotypes, which could have an impact on tumor biology.


Biochemical and Biophysical Research Communications | 2010

Abba promotes PDGF-mediated membrane ruffling through activation of the small GTPase Rac1

Datong Zheng; Shuqiong Niu; Dan Yu; Xiaoguo H. Zhan; Xianchun Zeng; Bota Cui; Yanping Chen; Jennifer R. Yoon; Stuart S. Martin; Xiang Lu; Xi Zhan

Abba is a member of the I-BAR-domain protein family that is characterized by a convex-shaped membrane-binding motif. Overexpression of GFP-tagged Abba in murine fibroblasts potentiated PDGF-mediated formation of membrane ruffles and lamellipodia. Immunofluorescent microscopy and pull-down analysis revealed that GFP-Abba colocalized with an active form of Rac1 in the membrane ruffles and enhanced the Rac GTPase activity in response to PDGF stimulation. Further immunoprecipitation assays demonstrated that GFP-Abba bound to both wild-type and constitutively active Rac1 and that the binding to either of the Rac1 forms was significantly enhanced upon PDGF stimulation. On the other hand, an Abba mutant deficient in Rac1 binding failed to promote membrane ruffling and Rac1 activation in response to PDGF. However, the cells overexpressing a truncated mutant carrying the I-BAR domain alone displayed numerous filopodia-like microspikes in a manner independent of growth factors. Also, the Rac-binding activity of the mutant was not affected by PDGF treatment. Our data indicates that the interaction between full-length Abba and Rac1 is implicated in membrane deformation and subjected to a growth factor-mediated regulation through the C-terminal sequence.


Oncotarget | 2015

The combinatorial activation of the PI3K and Ras/MAPK pathways is sufficient for aggressive tumor formation, while individual pathway activation supports cell persistence

Keyata Thompson; Rebecca A. Whipple; Jennifer R. Yoon; Michael M. Lipsky; Monica Charpentier; Amanda E. Boggs; Kristi R. Chakrabarti; Lekhana Bhandary; Lindsay K. Hessler; Stuart S. Martin; Michele I. Vitolo

A high proportion of human tumors maintain activation of both the PI3K and Ras/MAPK pathways. In basal-like breast cancer (BBC), PTEN expression is decreased/lost in over 50% of cases, leading to aberrant activation of the PI3K pathway. Additionally, BBC cell lines and tumor models have been shown to exhibit an oncogenic Ras-like gene transcriptional signature, indicating activation of the Ras/MAPK pathway. To directly test how the PI3K and Ras/MAPK pathways contribute to tumorigenesis, we deleted PTEN and activated KRas within non-tumorigenic MCF-10A breast cells. Neither individual mutation was sufficient to promote tumorigenesis, but the combination promoted robust tumor growth in mice. However, in vivo bioluminescence reveals that each mutation has the ability to promote a persistent phenotype. Inherent in the concept of tumor cell dormancy, a stage in which residual disease is present but remains asymptomatic, viable cells with each individual mutation can persist in vivo during a period of latency. The persistent cells were excised from the mice and showed increased levels of the cell cycle arrest proteins p21 and p27 compared to the aggressively growing PTEN−/−KRAS(G12V) cells. Additionally, when these persistent cells were placed into growth-promoting conditions, they were able to re-enter the cell cycle and proliferate. These results highlight the potential for either PTEN loss or KRAS activation to promote cell survival in vivo, and the unique ability of the combined mutations to yield rapid tumor growth. This could have important implications in determining recurrence risk and disease progression in tumor subtypes where these mutations are common.


Cancer Research | 2015

Abstract 2161: Activation of the MAPK pathway in combination with PTEN loss leads to aggressive primary tumor formation

Keyata Thompson; Rebecca A. Whipple; Jennifer R. Yoon; Monica Charpentier; Amanda E. Boggs; Lekhana Bhandary; Kristi R. Chakrabarti; Stuart S. Martin; Michele I. Vitolo

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Breast cancer is the most frequent malignancy in women, and although many breast cancers are curable via surgery, approximately one quarter maintain a latent and insidious characteristic of slow growth. The loss of the tumor suppressor PTEN is associated with breast cancer stage, increased lymph node status, and disease-related death, and the high rate of loss in primary tumors suggests a potential role in initiation and/or progression of the disease. Additionally, a large fraction of breast tumors carry oncogenic mutations resulting in the hyper-activation of the MAPK/ERK cascade (20%-25% ErbB2, 5% KRAS, 2% BRAF, 1% HRAS, 1% NRAS). Hyperactivation of survival and growth pathways is considered a hallmark of many human carcinomas, including breast cancer. The overactivation of the PI3K pathway (PTEN loss) or the MAPK pathway could grant a cell the ability to circumvent inhibitory pathways. However, specific cellular alterations in human breast epithelium controlled by PTEN inactivation and/or Ras activation, which lead to early primary tumor formation, remain poorly defined. Since the current view of cancer is based on a “multi-hit” hypothesis where human cancers display a multitude of genetic and epigenetic changes, and a number of such alterations are required for tumor development, the loss of PTEN (activation of the PI3K pathway) and expression of activated K-Ras(V12) (activation of the MAPK pathway) may cooperate to promote tumorigenesis. We therefore tested the hypothesis that the activation of the MAPK pathway via activated Ras expression in a PTEN-negative background promotes tumorgenicity. Using the non-tumorigenic human mammary cells line, MCF-10A, we created MCF-10A PTEN-/- cells, MCF-10A KRas(V12) cells, and MCF-10A PTEN-/-KRas(V12) cells. We have found that each mutation, independently and collectively, greatly enhanced cellular survival and regrowth efficiency of nutrient deprived and suspended cells in vitro. Using bioluminescent mouse xenograft models, we have determined the cells with either PTEN loss or KRas(V12) expression maintain an increased persistence in vivo up to 5 weeks beyond that of the parental cells, and the combination of PTEN loss and KRas(V12) expression resulted large tumors within 4 weeks of initial injection. The combination of “one-hit” to the PI3K pathway and “one-hit” to the MAPK pathways synergized to result in aggressive tumor growth, while each individual mutation only lead to cellular persistence in vivo, a characteristic that may have been previously overlooked in less sensitive xenograft models, either without bioluminescence imaging or when transplanting less genetically stable tumor cells. Citation Format: Keyata N. Thompson, Rebecca A. Whipple, Jennifer R. Yoon, Monica S. Charpentier, Amanda E. Boggs, Lekhana Bhandary, Kristi R. Chakrabarti, Stuart S. Martin, Michele I. Vitolo. Activation of the MAPK pathway in combination with PTEN loss leads to aggressive primary tumor formation. [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 2161. doi:10.1158/1538-7445.AM2015-2161


Cancer Research | 2013

Abstract 3772: The role of microtentacles in the metastatic potential of breast tumor stem cells.

Monica Charpentier; Rebecca A. Bettes; Michele I. Vitolo; Amanda E. Boggs; Jana Slovic; Keyata Thompson; Lekhana Bhandary; Jennifer R. Yoon; Stuart S. Martin

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Breast cancer is the 2nd leading cause of cancer-related death among women; however, the majority of deaths arise as complications from metastasis, rather than the primary tumor. The cancer stem cell (CSC) hypothesis provides an explanation for the limited success of current therapies for metastatic breast cancer. CSCs are defined as a subpopulation of tumor-initiating cells with the stem cell-like characteristics of self-renewal and multipotency. CSCs have been shown to enter circulation and reach distal tissues, where they may remain cell-cycle arrested, and therefore resistant to conventional chemotherapeutics. Recent work in our lab has shown that circulating tumor cells use dynamic tubulin-based microtentacles (McTNs) to reattach to distant tissues, a critical step in metastasis. McTNs are novel cellular structures formed by epithelial cells when detached from the extracellular matrix and are increased in more metastatic breast cancer cell lines. Given the proposed metastatic efficiency of CSCs, we examined McTN incidence and function in mammary stem cells and breast cancer stem cells. Flow cytometry for the CSC markers CD44 and CD24 showed that breast tumor cell lines with increased CSC characteristics display higher McTN frequencies. Given this correlation, CD44 and CD24 immunofluorescence was used to separate human mammary epithelial (HMLE) cells into CSC and non-CSC subpopulations with flow cytometry. Stem-like HMLE cells (CD44hi/CD24lo) have increased McTN levels compared to non-CSC cells (CD44lo/CD24hi) from the same HMLE cell line. The CSC subpopulation also demonstrated increased cytoskeletal modifications that promote McTN formation, such as elevated vimentin expression and an increase in both the amount and the bundling of stabilized, detyrosinated tubulin. Vimentin and detyrosinated tubulin localize to McTNs in suspended stem-like HMLEs. The increased McTNs in stem-like HMLEs promote faster initial reattachment of suspended cells that is inhibited by the tubulin-directed drug, Colchicine, confirming a functional role for McTNs in stem cell reattachment. Moreover, live cell confocal microscopy demonstrates that McTNs participate in the structure of breast stem cell mammopsheres, extending between adjacent cells along cell-cell junctions. McTNs can be reduced by the breast CSC targeting agent Curcumin. These studies show that McTNs contribute to the metastatic potential of breast CSCs as well as the ability of mammary stem cells to form multicellular mammospheres. We anticipate that this work will clarify the molecular mechanisms underlying tubulin alterations in breast cancer stem cells and identify how these tubulin modifications may be targeted more specifically to reduce McTNs and the metastatic reattachment efficiency of breast cancer CSCs. Citation Format: Monica S. Charpentier, Rebecca A. Bettes, Michele I. Vitolo, Amanda E. Boggs, Jana Slovic, Keyata Thompson, Lekhana Bhandary, Jennifer Yoon, Stuart S. Martin. The role of microtentacles in the metastatic potential of breast tumor stem cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 3772. doi:10.1158/1538-7445.AM2013-3772


Cancer Research | 2011

Abstract 3122: PTEN loss disrupts the actin cortex to promote tubulin microtentacle formation and increased metastatic potential

Michele I. Vitolo; Amanda E. Boggs; Keyata Thompson; Rebecca A. Whipple; Jennifer R. Yoon; Jana Slovic; Monica Charpentier; Michael A. Matrone; Stuart S. Martin

Breast cancer is the most frequent malignancy in women, and although many breast cancers are curable via surgery, approximately one quarter maintain a latent and insidious characteristic of slow growth with early metastasis. The loss of the tumor suppressor PTEN has been associated with breast cancer stage, lymph node status, and disease-related death, and the high rate of loss in primary tumors suggests a potential role in initiation and/or progression of the disease. However, specific cellular alterations in human breast epithelium controlled by PTEN inactivation, which lead to an increased metastatic phenotype, remain poorly defined. We have recently determined that PTEN expression loss leads to the production of long, dynamic, tubulin-based membrane protrusions upon detachment, which increase in frequency, number and length per cell compared to their isogenic, PTEN-expressing parental counterparts. These novel structures, termed microtentacles (McTNs), are structurally distinct from classical actin-based extensions of adherent cells, persist for days in breast tumor lines that are resistant to anoikis, and aid in the reattachment to matrix or cell monolayers and homo- and heterotypic aggregation. McTNs form when the balance of cytoskeletal forces shifts. In order to control morphology, normal cells counteract the expansion of microtubules with tension from the actin cortex. However, altering the balance between microtubules and actin has serious implications for circulating tumor cells (CTCs) dissemination, as metastatically efficient CTCs have been observed to avoid shear-induced fragmentation by undergoing sphere-to-cylinder shape transformations within capillaries. We, therefore, tested the hypothesis that PTEN loss disrupts the actin cortex to allow the increased production of McTNs. We determined that suspended PTEN-null mammary epithelial cells maintained elevated activation of the PI3K/Akt and MAPK pathways and apoptotic resistance to cell rounding and matrix detatchment, but neither activated pathway was responsible for the increased McTNs in these cells. The McTNs produced in the PTEN-null cells aid in cell reattachment, spreading, and homotypic aggregation, and Western blot analysis has proven that the PTEN-null cells show reduced inactivation of cofilin, an actin-binding protein known to sever actin filaments. Thus, the actin cytoskeleton in the PTEN-null cells contains more depolymerized actin, weakening the actin cortex. The combination of apoptotic resistance, the weakening of the actin cortex, and enhanced McTN formation due to PTEN loss may have important consequences for facilitating tumor cell extravasation and efficient adherence in metastatic sites. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3122. doi:10.1158/1538-7445.AM2011-3122


Cancer Research | 2009

Metastasis-associated microtentacles are induced in detached and circulating breast tumor cells by expression of the microtubule-binding protein, Tau.

Michael A. Matrone; Rebecca A. Whipple; Eric M. Balzer; Edward H. Cho; Jennifer R. Yoon; Stuart S. Martin

CTRC-AACR San Antonio Breast Cancer Symposium: 2008 Abstracts Abstract #55 Detached human breast tumor cell lines produce microtentacles composed of a unique kinesin-dependent coordination of vimentin filaments and detyrosinated microtubules. Compelling in vivo studies show that a mechanism consistent with microtentacles is responsible for the attachment of circulating tumor cells to blood vessel walls. However, the molecular regulators of microtentacle extension have only begun to be defined. Increased expression of the microtubule-binding protein, Tau, is associated with poor patient outcome in breast cancer, but the mechanism by which Tau protein affects prognosis remains unclear. We report here that expression of Tau directly regulates the formation of metastasis-associated microtentacles in detached and circulating breast tumor cells. First, increased expression of Tau correlates with increased microtentacle frequency in a panel of human breast tumor cell lines. Endogenous Tau protein colocalizes with tubulin microtentacles in cells detached from extracellular matrix. Exogenous expression of Tau significantly increases microtentacle frequency in weakly invasive breast tumor cell lines. Tau-induced microtentacles are longer, thicker and more rigid than those in cell lines without Tau expression. Although there is increasing interest in targeting the actin cytoskeleton to reduce tumor cell motility and division, our results indicate that breast tumor cells with high Tau expression have particularly dramatic increases in microtentacles when treated with inhibitors of actin polymerization. Likewise, tubulin-stabilizing compounds, like paclitaxel, increase microtentacles and may be ill-advised in patients with high Tau expression. Indeed, clinical studies show that high Tau expression increases the likelihood of recurrence following paclitaxel treatment. We observed an enrichment of Tau in metastatic tumors compared to matched primary tumors. So while stabilizing microtubules and disrupting actin filaments can each decrease tumor cell growth, it is important to consider the effects of these treatments on Tau-expressing cells to avoid accidentally enhancing the metastatic potential of circulating tumor cells while targeting cell division. Citation Information: Cancer Res 2009;69(2 Suppl):Abstract nr 55.

Collaboration


Dive into the Jennifer R. Yoon'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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge