Network


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

Hotspot


Dive into the research topics where Ross Lake is active.

Publication


Featured researches published by Ross Lake.


Cancer Research | 2011

LPA Receptor Heterodimerizes with CD97 to Amplify LPA-Initiated RHO-Dependent Signaling and Invasion in Prostate Cancer Cells

Yvona Ward; Ross Lake; Juan Juan Yin; Christopher Heger; Mark Raffeld; Paul Goldsmith; Maria J. Merino; Kathleen A. Kelly

CD97, an adhesion-linked G-protein-coupled receptor (GPCR), is induced in multiple epithelial cancer lineages. We address here the signaling properties and the functional significance of CD97 expression in prostate cancer. Our findings show that CD97 signals through Gα12/13 to increase RHO-GTP levels. CD97 functioned to mediate invasion in prostate cancer cells, at least in part, by associating with lysophosphatidic acid receptor 1 (LPAR1), leading to enhanced LPA-dependent RHO and extracellular signal-regulated kinase activation. Consistent with its role in invasion, depletion of CD97 in PC3 cells resulted in decreased bone metastasis without affecting subcutaneous tumor growth. Furthermore, CD97 heterodimerized and functionally synergized with LPAR1, a GPCR implicated in cancer progression. We also found that CD97 and LPAR expression were significantly correlated in clinical prostate cancer specimens. Taken together, these findings support the investigation of CD97 as a potential therapeutic cancer target.


PLOS ONE | 2012

TMPRSS2- Driven ERG Expression In Vivo Increases Self-Renewal and Maintains Expression in a Castration Resistant Subpopulation

Orla Casey; Lei Fang; Paul G. Hynes; Wassim Abou-Kheir; Philip Martin; Heather Tillman; Gyorgy Petrovics; Hibah O. Awwad; Yvona Ward; Ross Lake; Luhua Zhang; Kathleen Kelly

Genomic rearrangements commonly occur in many types of cancers and often initiate or alter the progression of disease. Here we describe an in vivo mouse model that recapitulates the most frequent rearrangement in prostate cancer, the fusion of the promoter region of TMPRSS2 with the coding region of the transcription factor, ERG. A recombinant bacterial artificial chromosome including an extended TMPRSS2 promoter driving genomic ERG was constructed and used for transgenesis in mice. TMPRSS2-ERG expression was evaluated in tissue sections and FACS-fractionated prostate cell populations. In addition to the anticipated expression in luminal cells, TMPRSS2-ERG was similarly expressed in the Sca-1hi/EpCAM+ basal/progenitor fraction, where expanded numbers of clonogenic self-renewing progenitors were found, as assayed by in vitro sphere formation. These clonogenic cells increased intrinsic self renewal in subsequent generations. In addition, ERG dependent self-renewal and invasion in vitro was demonstrated in prostate cell lines derived from the model. Clinical studies have suggested that the TMPRSS2-ERG translocation occurs early in prostate cancer development. In the model described here, the presence of the TMPRSS2-ERG fusion alone was not transforming but synergized with heterozygous Pten deletion to promote PIN. Taken together, these data suggest that one function of TMPRSS2-ERG is the expansion of self-renewing cells, which may serve as targets for subsequent mutations. Primary prostate epithelial cells demonstrated increased post transcriptional turnover of ERG compared to the TMPRSS2-ERG positive VCaP cell line, originally isolated from a prostate cancer metastasis. Finally, we determined that TMPRSS2-ERG expression occurred in both castration-sensitive and resistant prostate epithelial subpopulations, suggesting the existence of androgen-independent mechanisms of TMPRSS2 expression in prostate epithelium.


Infection and Immunity | 2007

Improved Antibacterial Host Defense and Altered Peripheral Granulocyte Homeostasis in Mice Lacking the Adhesion Class G Protein Receptor CD97

Tao Wang; Linhua Tian; Makoto Haino; Ji-Liang Gao; Ross Lake; Yvona Ward; Hongshan Wang; Ulrich Siebenlist; Philip M. Murphy; Kathleen Kelly

ABSTRACT CD97 is a member of the adhesion family of G protein-coupled receptors. Alternatively spliced forms of CD97 bind integrins α5β1 and αvβ3, decay accelerating factor, or dermatan sulfate. CD97 is expressed on myeloid cells at high levels and a variety of other cell types at lower levels. Little is known about the physiological function of CD97. To begin dissecting the function of CD97, we evaluated the immune response of CD97 null mice to systemic infection by Listeria monocytogenes. CD97 null mice were significantly more resistant to listeriosis than matched wild-type mice. A major determinant of the difference in survival appeared to be the comparatively more robust accumulation of granulocytes in the blood and in infected livers of CD97 null mice within 18 h of inoculation, correlating with a decrease in the number of bacteria. CD97 null mice also displayed a mild granulocytosis in the nonchallenged state. Because there is a strong suggestion that CD97 functions in an adhesive capacity, we examined the migratory properties of granulocytes in CD97 null mice. In chimeric animals, CD97 null and wild-type granulocytes migrated similarly, as determined by inflammation-induced emigration from the bone marrow and accumulation in the peritoneum. Granulocyte development in the bone marrow of CD97 null mice was comparable to that of wild-type mice, and CD97 deficiency did not appear to stimulate granulocytosis secondary to peripheral inflammation and resultant granulocyte colony-stimulating factor induction, unlike various other models of adhesion deficiencies. Our results suggest that CD97 plays a role in peripheral granulocyte homeostasis.


Cancer Research | 2014

AR-regulated TWEAK-FN14 pathway promotes prostate cancer bone metastasis.

Juan Juan Yin; Yen-Nien Liu; Heather Tillman; Ben Barrett; Stephen M. Hewitt; Kris Ylaya; Lei Fang; Ross Lake; Eva Corey; Colm Morrissey; Robert L. Vessella; Kathleen Kelly

The recurrence of prostate cancer metastases to bone after androgen deprivation therapy is a major clinical challenge. We identified FN14 (TNFRSF12A), a TNF receptor family member, as a factor that promotes prostate cancer bone metastasis. In experimental models, depletion of FN14 inhibited bone metastasis, and FN14 could be functionally reconstituted with IKKβ-dependent, NFκB signaling activation. In human prostate cancer, upregulated FN14 expression was observed in more than half of metastatic samples. In addition, FN14 expression was correlated inversely with androgen receptor (AR) signaling output in clinical samples. Consistent with this, AR binding to the FN14 enhancer decreased expression. We show here that FN14 may be a survival factor in low AR output prostate cancer cells. Our results define one upstream mechanism, via FN14 signaling, through which the NFκB pathway contributes to prostate cancer metastasis and suggest FN14 as a candidate therapeutic and imaging target for castrate-resistant prostate cancers.


Cell Reports | 2015

Identification of Different Classes of Luminal Progenitor Cells within Prostate Tumors

Supreet Agarwal; Paul G. Hynes; Heather Tillman; Ross Lake; Wassim Abou-Kheir; Lei Fang; Orla Casey; Amir H. Ameri; Philip Martin; Juan Juan Yin; Phillip J. Iaquinta; Wouter R. Karthaus; Hans Clevers; Charles L. Sawyers; Kathleen Kelly

Primary prostate cancer almost always has a luminal phenotype. However, little is known about the stem/progenitor properties of transformed cells within tumors. Using the aggressive Pten/Tp53-null mouse model of prostate cancer, we show that two classes of luminal progenitors exist within a tumor. Not only did tumors contain previously described multipotent progenitors, but also a major population of committed luminal progenitors. Luminal cells, sorted directly from tumors or grown as organoids, initiated tumors of adenocarcinoma or multilineage histological phenotypes, which is consistent with luminal and multipotent differentiation potentials, respectively. Moreover, using organoids we show that the ability of luminal-committed progenitors to self-renew is a tumor-specific property, absent in benign luminal cells. Finally, a significant fraction of luminal progenitors survived in vivo castration. In all, these data reveal two luminal tumor populations with different stem/progenitor cell capacities, providing insight into prostate cancer cells that initiate tumors and can influence treatment response.


PLOS ONE | 2011

Self-Renewing Pten-/-TP53-/- Protospheres Produce Metastatic Adenocarcinoma Cell Lines with Multipotent Progenitor Activity

Wassim Abou-Kheir; Paul G. Hynes; Philip Martin; Juan Juan Yin; Yen Nien Liu; Victoria Seng; Ross Lake; Joshua Spurrier; Kathleen Kelly

Prostate cancers of luminal adenocarcinoma histology display a range of clinical behaviors. Although most prostate cancers are slow-growing and indolent, a proportion is aggressive, developing metastasis and resistance to androgen deprivation treatment. One hypothesis is that a portion of aggressive cancers initiate from stem-like, androgen-independent tumor-propagating cells. Here we demonstrate the in vitro creation of a mouse cell line, selected for growth as self-renewing stem/progenitor cells, which manifests many in vivo properties of aggressive prostate cancer. Normal mouse prostate epithelium containing floxed Pten and TP53 alleles was subjected to CRE-mediated deletion in vitro followed by serial propagation as protospheres. A polyclonal cell line was established from dissociated protospheres and subsequently a clonal daughter line was derived. Both lines demonstrate a mature luminal phenotype in vitro. The established lines contain a stable minor population of progenitor cells with protosphere-forming ability and multi-lineage differentiation capacity. Both lines formed orthotopic adenocarcinoma tumors with metastatic potential to lung. Intracardiac inoculation resulted in brain and lung metastasis, while intra-tibial injection induced osteoblastic bone formation, recapitulating the bone metastatic phenotype of human prostate cancer. The cells showed androgen receptor dependent growth in vitro. Importantly, in vivo, the deprivation of androgens from established orthotopic tumors resulted in tumor regression and eventually castration-resistant growth. These data suggest that transformed prostate progenitor cells preferentially differentiate toward luminal cells and recapitulate many characteristics of the human disease.


Cancer Research | 2018

Abstract B018: A high-throughput screen identifies HSP90 inhibitors as potent therapeutics across multiple clinically representative organoid models of advanced prostate cancer

Keith H. Jansson; John Tucker; Lauren Stahl; John K. Simmons; Caitlyn Fuller; Michael L. Beshiri; Supreet Agarwal; Yasmine Abbey; Lei Fang; Paul G. Hynes; Alilin Aian Neil; Jacob Cawley; Ross Lake; Crystal Tran; Caitlin M. Tice; JuanJuan Yin; Xiahu Zhang; Rajarshi Guha; Shelley Hoover; R. Mark Simpson; Holly M. Nguyen; Eva Corey; Craig J. Thomas; David A. Proia; Kathleen A. Kelly

Androgen-deprivation therapy (ADT) remains the gold-standard therapy for prostate cancer (PrCa), and although ADT is initially effective, most men progress to castrate-resistant prostate cancer (CRPC) within 2-3 years. Advanced CRPC is challenging to treat because intrinsic tumor heterogeneity and phenotypic plasticity engender short-lived responses and underlie resistance to conventional therapies. Combined PTEN/TP53 alterations represent a major genotype of advanced CRPC (25-30%) and are associated with poor clinical outcomes. Established PrCa cell lines do not accurately represent the heterogeneity of advanced CRPC, and therefore, nonbiased pharmacogenomics screens have not been done. The development of clinically representative, tractable models suitable for high-throughput target identification and validation is crucial for advancing novel CRPC therapies to the clinic. A comprehensive nonbiased high-throughput screen performed on seven cell lines derived from a genetically engineered mouse model (GEMM) of Pten/Tp53 null PrCa identified strongly active compounds, including inhibitors of PI3K/AKT/mTOR signaling, the proteasome, cell cycle regulatory proteins, heat shock proteins, DNA repair signaling, NFKB signaling, MAPK signaling, and several types of epigenetic modifiers. HSP90 inhibitors were one of the most efficacious classes of compounds in the screen, and ganetespib, a clinically used second-generation HSP90 inhibitor with a favorable safety profile, was the most potent. Although HSP90 inhibitors have yet to be successful as single agents, they have not been thoroughly investigated in clinically representative models of advanced PrCa and have shown potential as “network drugs,” prompting our investigations into their utility in polytherapy. We first validated ganetespib as a single agent, where it displayed strong activity against several GEMM-derived and LuCaP PDX-derived organoid models encompassing genotypic, phenotypic, and lineage heterogeneity. These 10 novel LuCaP PDX-derived organoids are representative of the numerous categories of CRPC, including adenocarcinomas with wild-type AR, adenocarcinomas with altered AR, adenocarcinoma with neuroendocrine features, and neuroendocrine disease. Single-agent ganetespib was also strongly inhibitory in vivo, decreasing growth of Pten/Tp53 null endogenous GEMM tumors as well as a human PDX tumor. Mechanistic interrogation of cell lines, organoids, and tumors exposed to ganetespib revealed inhibition of targets from several inter-related networks including AR and pAKT, two central and mutually compensatory growth and survival pathways for PrCa. The efficacy of ganetespib against a diverse group of CRPC organoids and the simultaneous inhibition of PrCa survival signaling suggested it may work well in combination. We performed a proof-of-principle high-throughput matrix screen on organoids derived from a Pten/Tp53 null GEMM and identified docetaxel and etoposide to be synergistic when combined with ganetespib. Preclinical in vivo studies to validate these findings are ongoing. In all, comprehensive data from multiple near-patient models suggest novel contexts for second-generation HSP90-directed intervention against a variety of CRPC genotypes and phenotypes and expand upon the potential of HSP90 inhibitors to simultaneously inhibit oncogenic signaling and compensatory resistance mechanisms. Citation Format: Keith H. Jansson, John B. Tucker, Lauren E. Stahl, John K. Simmons, Caitlyn Fuller, Michael L. Beshiri, Supreet Agarwal, Yasmine Abbey, Lei Fang, Paul G. Hynes, Alilin Aian Neil, Jacob Cawley, Ross Lake, Crystal Tran, Caitlin M. Tice, JuanJuan Yin, Xiahu Zhang, Rajarshi Guha, Shelley Hoover, R. Mark Simpson, Holly Nguyen, Eva Corey, Craig J. Thomas, David Proia, Kathleen Kelly. A high-throughput screen identifies HSP90 inhibitors as potent therapeutics across multiple clinically representative organoid models of advanced prostate cancer [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr B018.


Cancer Research | 2016

Abstract LB-281: Elucidating potential therapeutic targets in a model of Pten/Tp53 null prostate cancer using high-throughput screening technology

Keith H. Jansson; John K. Simmons; Caitlyn Fuller; Supreet Agarwal; Paul G. Hynes; Lei Fang; Ross Lake; Jacob Cawley; Lauren Stahl; Xiaohu Zhang; Rajarshi Guha; Craig J. Thomas; Kathleen A. Kelly

Prostate cancer (PCa) demonstrates significant intra and inter tumor heterogeneity, similar to many other solid tumors. The “gold standard” of therapy for metastatic prostate cancer is androgen deprivation therapy (ADT). Although ADT is initially effective, acquired resistance, termed castration resistant prostate cancer (CRPC), almost always occurs. It is believed that development of CRPC is governed by both intrinsically resistant progenitor cells and by acquired genomic mutations. Clinically advanced CRPC tumors often display loss of PTEN (40%) and aberrations in TP53 (50%), the presence of which in primary prostate cancer are associated with poor clinical prognosis. Our goal is to identify novel targeted therapies against progenitor cells harboring Pten/Tp53 mutations. To enable high throughput screening, we derived multiple cell lines from a PB-Cre4 Ptenfl/fl;Tp53fl/fl mouse model before (intact) and after (castrate) castration. The RNA-seq analyses of the 8 resultant cell lines revealed distinct clustering of intact and castrate lines as well as patterns of expression characteristic of progenitor cells. Cell lineage marker analysis showed strong KRT8 and minimal KRT5 expression. Using these 8 distinct luminal cell lines, we performed the mechanism interrogation plate (MIPe) screen, a high throughput 1,912 compound screening assay at the National Center for Advancing Translational Science (NCATS). Post-screen informatics data processing and subsequent analysis sorted out compounds that displayed strong activity (1.1 and 1.2 curve class). Multiple compounds were unique to each cell line, however, no difference in sensitivity between the two groups of intact and castrate cell types was observed. Grouping of intact and castrate cell lines together as one population identified a diverse array of 235 compounds (12% of MIPe library) with robust activity against most of the cell lines. Some of these compounds are redundant for specific targets that are key regulators of a multitude of signaling pathways, such as the heat shock protein HSP90AB1, which was targeted by 11 different compounds. In contrast, other targets, like BIRC5, were potently targeted by only one compound. A majority of the 235 compounds targeted components of signaling pathways important in PCa, including: 28 compounds targeting PI3K/AKT/mTOR signaling (AKT1, PIK3CA, mTORc 1/2), 13 compounds targeting cell cycle regulators (CDK1, CDK4), 13 compounds targeting DNA repair and replication (CHEK1, TOP2A), 6 compounds targeting NFKB signaling (IKBKB,ITK) and 5 compounds targeting MAPK signaling (MAPK8, MAP2K1). Using high throughput screening technology to sort for compounds based on potency and activity identified clinically and biologically relevant therapeutic applications for existing compounds in a novel context, Pten/Tp53 null PCa progenitor cells. Citation Format: Keith H. Jansson, John K. Simmons, Caitlyn Fuller, Supreet Agarwal, Paul G. Hynes, Lei Fang, Ross Lake, Jacob Cawley, Lauren Stahl, Xiaohu Zhang, Rajarshi Guha, Craig Thomas, Kathleen Kelly. Elucidating potential therapeutic targets in a model of Pten/Tp53 null prostate cancer using high-throughput screening technology. [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 LB-281.


Cancer Research | 2013

Abstract 2708: The TWEAK-FN14 pathway promotes prostate cancer bone metastasis through the NFκB pathway.

Juan Juan Yin; Yen-Nien Liu; Ben Barrett; Lei Fang; Ross Lake; Orla Casey; Heather Tillman; Yvona Ward; Kathleen A. Kelly

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Advanced prostate cancer is highly associated with castration resistance and the development of bone metastases. Understanding the mechanisms contributing to prostate cancer bone metastasis is needed in order to develop novel therapeutic agents. NFκB signaling has been implicated in prostate cancer progression. The Tweak-Fn14 axis, which activates both canonical and non-canonical NFκB pathways, has been shown to be up-regulated in many cancer types. However, the role of the Tweak-Fn14 signaling in prostate cancer progression has not been investigated. In this study, we show that oncogenic Ras transformation of the AR negative, DU145 cell line led to the acquisition of bone metastatic capability and associated increased expression of TWEAK and FN14. Knocking down FN14 using shRNAs, as well as blocking the NFκB pathway using an IKBα super repressor, in DU145/Rasb1 cells, significantly inhibited bone metastasis and improved survival. This inhibitory effect of FN14shRNA was fully rescued by activating the NFκB canonical pathway, but not the NFκB non-canonical pathway. FN14 expression is also high in the bone metastatic, AR negative prostate cancer PC3 cell line. Similarly, knocking-down FN14 in PC3 cells inhibited bone metastatic capacity following intracardiac inoculation in a xenograft model. TWEAK and FN14 are relatively low or undetectable in AR positive prostate cancer cell lines including Lncap and 22RV1. AR binding was detected in both TWEAK and FN14 promoters as determined by chromatin immunoprecipitation analysis in Lncap. Reporter assays demonstrated that AR binding inhibited FN14 and TWEAK transcription. The above experimental models were further supported by analyses of publically available expression data sets for clinical prostate cancer samples that showed FN14 expression was inversely correlated with AR gene expression signatures. In addition, individual FN14 RNA mean expression was higher in clinical metastasis of castrate (n=11) as compared to non-castrate (n=8) patients. We propose that down-regulation of AR activity under castrate conditions may lead to increased FN14 expression and NFκB activation, which provides a survival benefit for prostate cancer cells associated with disease progression. Therefore, targeting the TWEAK-FN14 pathway in prostate cancer patients provides potential, new preventive and therapeutic approaches. Citation Format: Juan Juan Yin, Yen-Nien Liu, Ben Barrett, Lei Fang, Ross Lake, Orla Casey, Heather Tillman, Yvona Ward, Kathleen Kelly. The TWEAK-FN14 pathway promotes prostate cancer bone metastasis through the NFκB pathway. [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 2708. doi:10.1158/1538-7445.AM2013-2708


Blood | 2005

CD97, an adhesion receptor on inflammatory cells, stimulates angiogenesis through binding integrin counterreceptors on endothelial cells

Tao Wang; Yvona Ward; Linhua Tian; Ross Lake; Liliana Guedez; William G. Stetler-Stevenson; Kathleen A. Kelly

Collaboration


Dive into the Ross Lake's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lei Fang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Paul G. Hynes

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yvona Ward

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Juan Juan Yin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Heather Tillman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Philip Martin

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Wassim Abou-Kheir

American University of Beirut

View shared research outputs
Top Co-Authors

Avatar

Kathleen Kelly

American University of Beirut

View shared research outputs
Top Co-Authors

Avatar

Craig J. Thomas

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge