Network


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

Hotspot


Dive into the research topics where Isla P. Garraway is active.

Publication


Featured researches published by Isla P. Garraway.


Science | 2010

Identification of a cell-of-origin for human prostate cancer

Andrew S. Goldstein; Jiaoti Huang; Changyong Guo; Isla P. Garraway; Owen N. Witte

Another Cell Culprit in Prostate Cancer A recent controversial hypothesis about the cellular origins of human cancer, the so-called “cancer stem cell hypothesis,” has fueled interest in identifying the specific cell types that give rise to common epithelial cancers. A single, well-defined cell of origin could, in principle, lead to more effective targeted therapies. Based on histological evidence and/or studies of mouse tumors, luminal cells are believed to be the cell of origin in prostate cancer. Now, using functional assays of cells derived from benign human prostate tissue, Goldstein et al. (p. 568) find that a different cell type, basal cells, can give rise in mice to prostate tumors that closely resemble human prostate tumors. Thus, the cellular origin of prostate cancer may be more complex than anticipated. A new experimental model identifies basal cells, rather than luminal cells, as the origin of prostate cancer. Luminal cells are believed to be the cells of origin for human prostate cancer, because the disease is characterized by luminal cell expansion and the absence of basal cells. Yet functional studies addressing the origin of human prostate cancer have not previously been reported because of a lack of relevant in vivo human models. Here we show that basal cells from primary benign human prostate tissue can initiate prostate cancer in immunodeficient mice. The cooperative effects of AKT, ERG, and androgen receptor in basal cells recapitulated the histological and molecular features of human prostate cancer, with loss of basal cells and expansion of luminal cells expressing prostate-specific antigen and alpha-methylacyl-CoA racemase. Our results demonstrate that histological characterization of cancers does not necessarily correlate with the cellular origins of the disease.


Cancer Cell | 2011

Cell Autonomous Role of PTEN in Regulating Castration-Resistant Prostate Cancer Growth

David J. Mulholland; Linh M. Tran; Yunfeng Li; Houjian Cai; Ashkan Morim; Shunyou Wang; Seema Plaisier; Isla P. Garraway; Jiaoti Huang; Thomas G. Graeber; Hong Wu

Alteration of the PTEN/PI3K pathway is associated with late-stage and castrate-resistant prostate cancer (CRPC). However, how PTEN loss is involved in CRPC development is not clear. Here, we show that castration-resistant growth is an intrinsic property of Pten null prostate cancer (CaP) cells, independent of cancer development stage. PTEN loss suppresses androgen-responsive gene expressions by modulating androgen receptor (AR) transcription factor activity. Conditional deletion of Ar in the epithelium promotes the proliferation of Pten null cancer cells, at least in part, by downregulating the androgen-responsive gene Fkbp5 and preventing PHLPP-mediated AKT inhibition. Our findings identify PI3K and AR pathway crosstalk as a mechanism of CRPC development, with potentially important implications for CaP etiology and therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Trop2 identifies a subpopulation of murine and human prostate basal cells with stem cell characteristics

Andrew S. Goldstein; Devon A. Lawson; Donghui Cheng; Wenyi Sun; Isla P. Garraway; Owen N. Witte

The epithelium of the adult prostate contains 3 distinct cell types: basal, luminal, and neuroendocrine. Tissue-regenerative activity has been identified predominantly from the basal cells, isolated by expression of CD49f and stem cell antigen-1 (Sca-1). An important question for the field is whether all basal cells have stem cell characteristics. Prostate-specific microarray databases were interrogated to find candidate surface antigens that could subfractionate the basal cell population. Tumor-associated calcium signal transducer 2 (TACSTD2/Trop2/M1S1/GA733-1) was identified because it was enriched after castration, in prostate sphere cells and in the basal fraction. In the murine prostate, Trop2 shows progenitor characteristics such as localization to the region of the gland proximal to the urethra and enrichment for sphere-forming and colony-forming cells. Trop2 subfractionates the basal cells into 2 populations, both of which express characteristic basal cell markers by quantitative PCR. However, only the basal cells expressing high levels of Trop2 were able to efficiently form spheres in vitro. In the human prostate, where Sca-1 is not expressed, sphere-forming progenitor cells were also isolated based on high expression of Trop2 and CD49f. Trop2-expressing murine basal cells could regenerate prostatic tubules in vivo, whereas the remaining basal cells had minimal activity. Evidence was found for basal, luminal, and neuroendocrine cells in prostatic tubules regenerated from Trop2hi basal cells. In summary, functionally distinct populations of cells exist within the prostate basal compartment and an epithelial progenitor can give rise to neuroendocrine cells in vivo.


The Prostate | 2009

Human prostate sphere-forming cells represent a subset of basal epithelial cells capable of glandular regeneration in vivo.

Isla P. Garraway; Wenyi Sun; Chau P. Tran; Sven Perner; Bao Zhang; Andrew S. Goldstein; Scott Hahm; Maahum Haider; Christian Head; Robert E. Reiter; Mark A. Rubin; Owen N. Witte

Prostate stem/progenitor cells function in glandular development and maintenance. They may be targets for tumor initiation, so characterization of these cells may have therapeutic implications. Cells from dissociated tissues that form spheres in vitro often represent stem/progenitor cells. A subset of human prostate cells that form prostaspheres were evaluated for self‐renewal and tissue regeneration capability in the present study.


PLOS ONE | 2012

Identification of CD166 as a Surface Marker for Enriching Prostate Stem/Progenitor and Cancer Initiating Cells

Jing Jiao; Antreas Hindoyan; Shunyou Wang; Linh M. Tran; Andrew S. Goldstein; Devon A. Lawson; Donghui Chen; Yunfeng Li; Changyong Guo; Baohui Zhang; Ladan Fazli; Martin Gleave; Owen N. Witte; Isla P. Garraway; Hong Wu

New therapies for late stage and castration resistant prostate cancer (CRPC) depend on defining unique properties and pathways of cell sub-populations capable of sustaining the net growth of the cancer. One of the best enrichment schemes for isolating the putative stem/progenitor cell from the murine prostate gland is Lin-;Sca1+;CD49fhi (LSChi), which results in a more than 10-fold enrichment for in vitro sphere-forming activity. We have shown previously that the LSChi subpopulation is both necessary and sufficient for cancer initiation in the Pten-null prostate cancer model. To further improve this enrichment scheme, we searched for cell surface molecules upregulated upon castration of murine prostate and identified CD166 as a candidate gene. CD166 encodes a cell surface molecule that can further enrich sphere-forming activity of WT LSChi and Pten null LSChi. Importantly, CD166 could enrich sphere-forming ability of benign primary human prostate cells in vitro and induce the formation of tubule-like structures in vivo. CD166 expression is upregulated in human prostate cancers, especially CRPC samples. Although genetic deletion of murine CD166 in the Pten null prostate cancer model does not interfere with sphere formation or block prostate cancer progression and CRPC development, the presence of CD166 on prostate stem/progenitors and castration resistant sub-populations suggest that it is a cell surface molecule with the potential for targeted delivery of human prostate cancer therapeutics.


Journal of Biological Chemistry | 2012

Secreted Hsp90 Is a Novel Regulator of the Epithelial to Mesenchymal Transition (EMT) in Prostate Cancer

Michael W. Hance; Krystal Dole; Udhayakumar Gopal; Jessica E. Bohonowych; Agnieszka Jezierska-Drutel; Carola A. Neumann; Haibo Liu; Isla P. Garraway; Jennifer S. Isaacs

Background: Epithelial to mesenchymal transition (EMT) correlates with increased metastatic potential and poor prognosis. Results: Secreted eHsp90 induces EMT, matrix metalloproteinase activity and cell motility. Conclusion: EMT inducing activity of eHsp90 provides a mechanistic basis for its tumorigenic and metastatic function. Significance: The requirement for eHsp90 in supporting tumorigenic events indicates that targeting eHsp90 may represent a therapeutic approach to improve prostate cancer patient survival. Prostate cancer (PCa) is the most frequently diagnosed malignancy in men, and the second highest contributor of male cancer related lethality. Disease mortality is due primarily to metastatic spread, highlighting the urgent need to identify factors involved in this progression. Activation of the genetic epithelial to mesenchymal transition (EMT) program is implicated as a major contributor of PCa progression. Initiation of EMT confers invasive and metastatic behavior in preclinical models and is correlated with poor clinical prognosis. Extracellular Hsp90 (eHsp90) promotes cell motility and invasion in cancer cells and metastasis in preclinical models, however, the mechanistic basis for its widespread tumorigenic function remains unclear. We have identified a novel and pivotal role for eHsp90 in driving EMT events in PCa. In support of this notion, more metastatic PCa lines exhibited increased eHsp90 expression relative to their lineage-related nonmetastatic counterparts. We demonstrate that eHsp90 promoted cell motility in an ERK and matrix metalloproteinase-2/9-dependent manner, and shifted cellular morphology toward a mesenchymal phenotype. Conversely, inhibition of eHsp90 attenuated pro-motility signaling, blocked PCa migration, and shifted cell morphology toward an epithelial phenotype. Last, we report that surface eHsp90 was found in primary PCa tumor specimens, and elevated eHsp90 expression was associated with increased levels of matrix metalloproteinase-2/9 transcripts. We conclude that eHsp90 serves as a driver of EMT events, providing a mechanistic basis for its ability to promote cancer progression and metastasis in preclinical models. Furthermore, its newly identified expression in PCa specimens, and potential regulation of pro-metastatic genes, supports a putative clinical role for eHsp90 in PCa progression.


PLOS ONE | 2012

Epcam, CD44, and CD49f distinguish sphere-forming human prostate basal cells from a subpopulation with predominant tubule initiation capability.

Changyong Guo; Haibo Liu; Baohui Zhang; Radu M. Cadaneanu; Aqila Mayle; Isla P. Garraway

Background Human prostate basal cells expressing alpha-6 integrin (CD49fHi) and/or CD44 form prostaspheres in vitro. This functional trait is often correlated with stem/progenitor (S/P) activity, including the ability to self-renew and induce differentiated tubules in vivo. Antigenic profiles that distinguish tubule-initiating prostate stem cells (SCs) from progenitor cells (PCs) and mature luminal cells (LCs) with less regenerative potential are unknown. Methodology/Principle Findings Prostasphere assays and RT-PCR analysis was performed following FACS separation of total benign prostate cells based upon combinations of Epcam, CD44, and/or CD49f expression. Epithelial cell fractions were isolated, including Epcam+CD44+ and Epcam+CD44+CD49fHi basal cells that formed abundant spheres. When non-sphere-forming Epcam+CD44− cells were fractionated based upon CD49f expression, a distinct subpopulation (Epcam+CD44−CD49fHi) was identified that possessed a basal profile similar to Epcam+CD44+CD49fHi sphere-forming cells (p63+ARLoPSA−). Evaluation of tubule induction capability of fractionated cells was performed, in vivo, via a fully humanized prostate tissue regeneration assay. Non-sphere-forming Epcam+CD44− cells induced significantly more prostate tubular structures than Epcam+CD44+ sphere-forming cells. Further fractionation based upon CD49f co-expression identified Epcam+CD44−CD49fHi (non-sphere-forming) basal cells with significantly increased tubule induction activity compared to Epcam+CD44−CD49fLo (true) luminal cells. Conclusions/Significance Our data delineates antigenic profiles that functionally distinguish human prostate epithelial subpopulations, including putative SCs that display superior tubule initiation capability and induce differentiated ductal/acini structures, sphere-forming PCs with relatively decreased tubule initiation activity, and terminally differentiated LCs that lack both sphere–forming and tubule-initiation activity. The results clearly demonstrate that sphere-forming ability is not predictive of tubule-initiation activity. The subpopulations identified are of interest because they may play distinct roles as cells of origin in the development of prostatic diseases, including cancer.


Urologic Oncology-seminars and Original Investigations | 2010

Phospho-Akt pathway activation and inhibition depends on N-cadherin or phospho-EGFR expression in invasive human bladder cancer cell lines

Hervé Wallerand; Ying Cai; Zev A. Wainberg; Isla P. Garraway; Isabelle Lascombe; Gaëlle Nicolle; Jean Paul Thiery; Hugues Bittard; François Radvanyi; Robert R. Reiter

OBJECTIVES A particular interest in epithelial-mesenchymal transition (EMT), which takes place during embryonic development, provided potential mechanisms involved in the progression of many epithelial tumors, including bladder cancer (BC). The phospho-Akt signaling pathway is supposed to be involved in invasion and progression of human tumors, including BC. Moreover, it has been demonstrated in bladder cancer cell lines that N-cadherin or phospho-epithelial growth factor receptor (EGFR) expression are correlated to tumor progression. Our objectives were to evaluate the potential phospho-Akt pathway involvement in N-cadherin and/or phospho-EGFR positive BC cell lines and to evaluate the prognostic value of E- and N-cadherin expression in patients undergoing cystectomy for invasive BC. MATERIALS AND METHODS We screened a panel of invasive and noninvasive BC cell lines for E- and N-cadherin, phospho-EGFR, and phospho-Akt expression using the Western blot technique (WB). The potential role of N-cadherin in invasion was assessed by Matrigel assays with and without the N-cadherin blocking monoclonal antibody GC-4. Then we used the Affymetrix microarray technique to evaluate the prognostic value of E- and N-cadherin expression in 30 patients undergoing a cystectomy for invasive BC. RESULTS N-cadherin and phospho-EGFR expression are associated with Akt activation and with invasive behavior modulation. Even if Akt activation is sufficient in promoting invasion, its inactivation by LY294002 (PI-3 kinase inhibitor) is less efficient on invasion than inhibition of N-cadherin and phospho-EGFR by GC-4 (monoclonal antibody) and gefitinib (anti-tyrosine kinase), respectively. N-cadherin and phospho-EGFR inhibition decreased phospho-Akt activation but also caused restoration and reinforcing of E-cadherin expression, respectively, while phospho-Akt inhibition did not have any impact on E-cadherin expression. In a group of high-risk bladder tumors (T(1)G(3)), N- and E-cadherin expression could be considered as a prognostic marker. In a group of patients with invasive BC (pT(2)-T(4)) undergoing cystectomy, we showed a shorter overall survival when BC expressed N-cadherin (P = 0.0064) and when E-cadherin expression was down-regulated (P = 0.00165). The N (positive) /E (negative) profile has the worst prognosis (P = 0.00153). CONCLUSIONS We confirmed the partial responsibility of p-Akt activation in invasion of some BC cell lines expressing N-cadherin or p-EGFR and also the potential role of N-cadherin and p-EGFR as target in cancer therapy. N/E- cadherin expression profile has a significant prognostic value in invasive BC.


The Journal of Urology | 2010

Absent CD44v6 Expression is an Independent Predictor of Poor Urothelial Bladder Cancer Outcome

Tobias Klatte; David Seligson; Jianyu Rao; Hong Yu; Michela de Martino; Isla P. Garraway; Steven G. Wong; Arie S. Belldegrun; Allan J. Pantuck

PURPOSE CD44v6 is a cell surface protein involved in cell migration, cell adhesion, tumor progression and metastatic spread. We evaluated its role as a molecular marker for urothelial bladder cancer. MATERIALS AND METHODS A tissue microarray was constructed containing 410 primary urothelial bladder cancers, each in triplicate. Immunohistochemical staining was done with a commercially available antibody. The percent of tumor cells staining positive for CD44v6 was evaluated and we assessed associations with stage, grade and survival. RESULTS CD44v6 expression was higher in noninvasive (Ta, Tis) vs invasive (T1-T4) tumors (p <0.001). It decreased with increasing grade (p <0.001). In patients who underwent transurethral bladder resection absent CD44v6 expression was associated with a 2.3-fold increased risk of recurrence (95% CI 1.28 to 4.08). Median time to recurrence for tumors with vs without CD44v6 expression was 23 vs 9 months (p = 0.003). In a multivariate Cox model absent CD44 expression was an independent adverse prognostic factor for tumor recurrence (HR 2.33, p = 0.006). In cystectomy cases median overall survival for CD44v6 nonexpression vs expression was 30 vs 75 months (p = 0.0027) and CD44v6 expression was retained as an independent prognostic factor for overall survival (HR 1.54, p = 0.042). CONCLUSIONS Absent CD44v6 expression is an independent adverse predictor of urothelial bladder cancer recurrence and overall survival. Routine evaluation of CD44v6 expression may allow the identification of high risk patients who require more intensive surveillance or aggressive therapy. Targeting of CD44v6 with monoclonal antibodies may provide new avenues for urothelial bladder cancer imaging and treatment.


Cancer Research | 2016

Integrated classification of prostate cancer reveals a novel luminal subtype with poor outcome

Sungyong You; Beatrice Knudsen; Nicholas Erho; Mohammed Alshalalfa; Mandeep Takhar; Hussam Al-Deen Ashab; Elai Davicioni; R. Jeffrey Karnes; Eric A. Klein; Robert B. Den; Ashley E. Ross; Edward M. Schaeffer; Isla P. Garraway; Jayoung Kim; Michael R. Freeman

Prostate cancer is a biologically heterogeneous disease with variable molecular alterations underlying cancer initiation and progression. Despite recent advances in understanding prostate cancer heterogeneity, better methods for classification of prostate cancer are still needed to improve prognostic accuracy and therapeutic outcomes. In this study, we computationally assembled a large virtual cohort (n = 1,321) of human prostate cancer transcriptome profiles from 38 distinct cohorts and, using pathway activation signatures of known relevance to prostate cancer, developed a novel classification system consisting of three distinct subtypes (named PCS1-3). We validated this subtyping scheme in 10 independent patient cohorts and 19 laboratory models of prostate cancer, including cell lines and genetically engineered mouse models. Analysis of subtype-specific gene expression patterns in independent datasets derived from luminal and basal cell models provides evidence that PCS1 and PCS2 tumors reflect luminal subtypes, while PCS3 represents a basal subtype. We show that PCS1 tumors progress more rapidly to metastatic disease in comparison with PCS2 or PCS3, including PSC1 tumors of low Gleason grade. To apply this finding clinically, we developed a 37-gene panel that accurately assigns individual tumors to one of the three PCS subtypes. This panel was also applied to circulating tumor cells (CTC) and provided evidence that PCS1 CTCs may reflect enzalutamide resistance. In summary, PCS subtyping may improve accuracy in predicting the likelihood of clinical progression and permit treatment stratification at early and late disease stages. Cancer Res; 76(17); 4948-58. ©2016 AACR.

Collaboration


Dive into the Isla P. Garraway's collaboration.

Top Co-Authors

Avatar

Beatrice Knudsen

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Baohui Zhang

University of California

View shared research outputs
Top Co-Authors

Avatar

Michael T. Lewis

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Owen N. Witte

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael R. Freeman

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sungyong You

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Changyong Guo

University of California

View shared research outputs
Top Co-Authors

Avatar

Eric T. Miller

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge