Network


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

Hotspot


Dive into the research topics where Samantha Morley is active.

Publication


Featured researches published by Samantha Morley.


American Journal of Pathology | 2012

Large Oncosomes in Human Prostate Cancer Tissues and in the Circulation of Mice with Metastatic Disease

Dolores Di Vizio; Matteo Morello; Andrew C. Dudley; Peter Schow; Rosalyn M. Adam; Samantha Morley; David J. Mulholland; Mirja Rotinen; Martin H. Hager; Luigi Insabato; Marsha A. Moses; Francesca Demichelis; Michael P. Lisanti; Hong Wu; Michael Klagsbrun; Neil A. Bhowmick; Mark A. Rubin; Crislyn D'Souza-Schorey; Michael R. Freeman

Oncosomes are tumor-derived microvesicles that transmit signaling complexes between cell and tissue compartments. Herein, we show that amoeboid tumor cells export large (1- to 10-μm diameter) vesicles, derived from bulky cellular protrusions, that contain metalloproteinases, RNA, caveolin-1, and the GTPase ADP-ribosylation factor 6, and are biologically active toward tumor cells, endothelial cells, and fibroblasts. We describe methods by which large oncosomes can be selectively sorted by flow cytometry and analyzed independently of vesicles <1 μm. Structures resembling large oncosomes were identified in the circulation of different mouse models of prostate cancer, and their abundance correlated with tumor progression. Similar large vesicles were also identified in human tumor tissues, but they were not detected in the benign compartment. They were more abundant in metastases. Our results suggest that tumor microvesicles substantially larger than exosome-sized particles can be visualized and quantified in tissues and in the circulation, and isolated and characterized using clinically adaptable methods. These findings also suggest a mechanism by which migrating tumor cells condition the tumor microenvironment and distant sites, thereby potentiating advanced disease.


Embo Molecular Medicine | 2012

DIAPH3 governs the cellular transition to the amoeboid tumour phenotype

Martin H. Hager; Samantha Morley; Diane R. Bielenberg; Sizhen Gao; Matteo Morello; Ilona N. Holcomb; Wennuan Liu; Ghassan Mouneimne; Francesca Demichelis; Jayoung Kim; Keith R. Solomon; Rosalyn M. Adam; William B. Isaacs; Henry N. Higgs; Robert L. Vessella; Dolores Di Vizio; Michael R. Freeman

Therapies for most malignancies are generally ineffective once metastasis occurs. While tumour cells migrate through tissues using diverse strategies, the signalling networks controlling such behaviours in human tumours are poorly understood. Here we define a role for the Diaphanous‐related formin‐3 (DIAPH3) as a non‐canonical regulator of metastasis that restrains conversion to amoeboid cell behaviour in multiple cancer types. The DIAPH3 locus is close to RB1, within a narrow consensus region of deletion on chromosome 13q in prostate, breast and hepatocellular carcinomas. DIAPH3 silencing in human carcinoma cells destabilized microtubules and induced defective endocytic trafficking, endosomal accumulation of EGFR, and hyperactivation of EGFR/MEK/ERK signalling. Silencing also evoked amoeboid properties, increased invasion and promoted metastasis in mice. In human tumours, DIAPH3 down‐regulation was associated with aggressive or metastatic disease. DIAPH3‐silenced cells were sensitive to MEK inhibition, but showed reduced sensitivity to EGFR inhibition. These findings have implications for understanding mechanisms of metastasis, and suggest that identifying patients with chromosomal deletions at DIAPH3 may have prognostic value.


Cancer Biology & Therapy | 2014

Enhanced shedding of extracellular vesicles from amoeboid prostate cancer cells: potential effects on the tumor microenvironment.

Jayoung Kim; Samantha Morley; Minh T. N. Le; Denis Bedoret; Dale T. Umetsu; Dolores Di Vizio; Michael R. Freeman

The gene encoding the cytoskeletal regulator DIAPH3 is lost at high frequency in metastatic prostate cancer, and DIAPH3 silencing evokes a transition to an amoeboid tumor phenotype in multiple cell backgrounds. This amoeboid transformation is accompanied by increased tumor cell migration, invasion, and metastasis. DIAPH3 silencing also promotes the formation of atypically large (>1 μm) membrane blebs that can be shed as extracellular vesicles (EV) containing bioactive cargo. Whether loss of DIAPH3 also stimulates the release of nano-sized EV (e.g., exosomes) is not established. Here we examined the mechanism of release and potential biological functions of EV shed from DIAPH3-silenced and other prostate cancer cells. We observed that stimulation of LNCaP cells with the prostate stroma-derived growth factor heparin-binding EGF-like growth factor (HB-EGF), combined with p38MAPK inhibition caused EV shedding, a process mediated by ERK1/2 hyperactivation. DIAPH3 silencing in DU145 cells also increased rates of EV production. EV isolated from DIAPH3-silenced cells activated AKT1 and androgen signaling, increased proliferation of recipient tumor cells, and suppressed proliferation of human macrophages and peripheral blood mononuclear cells. DU145 EV contained miR-125a, which suppressed AKT1 expression and proliferation in recipient human peripheral blood mononuclear cells and macrophages. Our findings suggest that EV produced as a result of DIAPH3 loss or growth factor stimulation may condition the tumor microenvironment through multiple mechanisms, including the proliferation of cancer cells and suppression of tumor-infiltrating immune cells.


Cell Communication and Signaling | 2014

Integration of proteomic and transcriptomic profiles identifies a novel PDGF-MYC network in human smooth muscle cells

Wei Yang; Sungyong You; Hyobin Jeong; Samantha Morley; Michelle Mulone; Tanya Logvinenko; Jayoung Kim; Daehee Hwang; Michael R. Freeman; Rosalyn M. Adam

BackgroundPlatelet-derived growth factor-BB (PDGF-BB) has been implicated in the proliferation, migration and synthetic activities of smooth muscle cells that characterize physiologic and pathologic tissue remodeling in hollow organs. However, neither the molecular basis of PDGFR-regulated signaling webs, nor the extent to which specific components within these networks could be exploited for therapeutic benefit has been fully elucidated.ResultsExpression profiling and quantitative proteomics analysis of PDGF-treated primary human bladder smooth muscle cells identified 1,695 genes and 241 proteins as differentially expressed versus non-treated cells. Analysis of gene expression data revealed MYC, JUN, EGR1, MYB, RUNX1, as the transcription factors most significantly networked with up-regulated genes. Forty targets were significantly altered at both the mRNA and protein levels. Proliferation, migration and angiogenesis were the biological processes most significantly associated with this signature, and MYC was the most highly networked master regulator. Alterations in master regulators and gene targets were validated in PDGF-stimulated smooth muscle cells in vitro and in a model of bladder injury in vivo. Pharmacologic inhibition of MYC and JUN confirmed their role in SMC proliferation and migration. Network analysis identified the diaphanous-related formin 3 as a novel PDGF target regulated by MYC and JUN, which was necessary for PDGF-stimulated lamellipodium formation.ConclusionsThese findings provide the first systems-level analysis of the PDGF-regulated transcriptome and proteome in normal smooth muscle cells. The analyses revealed an extensive cohort of PDGF-dependent biological processes and connected key transcriptional effectors to their regulation, significantly expanding current knowledge of PDGF-stimulated signaling cascades. These observations also implicate MYC as a novel target for pharmacological intervention in fibroproliferative expansion of smooth muscle, and potentially in cancers in which PDGFR-dependent signaling or MYC activation promote tumor progression.


Scientific Reports | 2015

Regulation of microtubule dynamics by DIAPH3 influences amoeboid tumor cell mechanics and sensitivity to taxanes

Samantha Morley; Sungyong You; Sara Pollan; Jiyoung Choi; Bo Zhou; Martin H. Hager; Kenneth Steadman; Cristiana Spinelli; Kavitha Rajendran; Arkadiusz Gertych; Jayoung Kim; Rosalyn M. Adam; Wei Yang; Ramaswamy Krishnan; Beatrice Knudsen; Dolores Di Vizio; Michael R. Freeman

Taxanes are widely employed chemotherapies for patients with metastatic prostate and breast cancer. Here, we show that loss of Diaphanous-related formin-3 (DIAPH3), frequently associated with metastatic breast and prostate cancers, correlates with increased sensitivity to taxanes. DIAPH3 interacted with microtubules (MT), and its loss altered several parameters of MT dynamics as well as decreased polarized force generation, contractility, and response to substrate stiffness. Silencing of DIAPH3 increased the cytotoxic response to taxanes in prostate and breast cancer cell lines. Analysis of drug activity for tubulin-targeted agents in the NCI-60 cell line panel revealed a uniform positive correlation between reduced DIAPH3 expression and drug sensitivity. Low DIAPH3 expression correlated with improved relapse-free survival in breast cancer patients treated with chemotherapeutic regimens containing taxanes. Our results suggest that inhibition of MT stability arising from DIAPH3 downregulation enhances susceptibility to MT poisons, and that the DIAPH3 network potentially reports taxane sensitivity in human tumors.


Asian Journal of Andrology | 2014

Trading in your spindles for blebs: the amoeboid tumor cell phenotype in prostate cancer

Samantha Morley; Martin H. Hager; Sara Pollan; Beatrice Knudsen; Dolores Di Vizio; Michael R. Freeman

Prostate cancer (PCa) remains a principal cause of mortality in developed countries. Because no clinical interventions overcome resistance to androgen ablation therapy, management of castration resistance and metastatic disease remains largely untreatable. Metastasis is a multistep process in which tumor cells lose cell-cell contacts, egress from the primary tumor, intravasate, survive shear stress within the vasculature and extravasate into tissues to colonize ectopic sites. Tumor cells reestablish migratory behaviors employed during nonneoplastic processes such as embryonic development, leukocyte trafficking and wound healing. While mesenchymal motility is an established paradigm of dissemination, an alternate, ‘amoeboid’ phenotype is increasingly appreciated as relevant to human cancer. Here we discuss characteristics and pathways underlying the phenotype, and highlight our findings that the cytoskeletal regulator DIAPH3 governs the mesenchymal-amoeboid transition. We also describe our identification of a new class of tumor-derived microvesicles, large oncosomes, produced by amoeboid cells and with potential clinical utility in prostate and other cancers.


Journal of Proteome Research | 2016

Universal Solid-Phase Reversible Sample-Prep for Concurrent Proteome and N-Glycome Characterization

Hui Zhou; Samantha Morley; Stephen Kostel; Michael R. Freeman; Vivek Joshi; David C. Brewster; Richard S. Lee

We describe a novel solid-phase reversible sample-prep (SRS) platform that enables rapid sample preparation for concurrent proteome and N-glycome characterization for nearly all protein samples. SRS utilizes a uniquely functionalized, silica-based bead that has strong affinity toward proteins with minimal to no affinity for peptides and other small molecules. By leveraging this inherent size difference between proteins and peptides, SRS permits high-capacity binding of proteins, rapid removal of small molecules (detergents, metabolites, salts, peptides, etc.), extensive manipulation including enzymatic and chemical treatments on bead-bound proteins, and easy recovery of N-glycans and peptides. SRS was evaluated in a wide range of samples including glycoproteins, cell lysate, murine tissues, and human urine. SRS was also coupled to a quantitative strategy to investigate the differences between DU145 prostate cancer cells and its DIAPH3-silenced counterpart. Previous studies suggested that DIAPH3 silencing in DU145 induced transition to an amoeboid phenotype that correlated with tumor progression and metastasis. In this pilot study we identified distinct proteomic and N-glycomic alterations between them. A metastasis-associated tyrosine kinase receptor ephrin-type-A receptor (EPHA2) was highly up-regulated in DIAPH3-silenced cells, indicating a possible connection between EPHA2 and DIAPH3. Moreover, distinct alterations in the N-glycome were identified, suggesting cross-links between DIAPH3 and glycosyltransferase networks.


Oncotarget | 2018

Distinct age-associated molecular profiles in acute myeloid leukemia defined by comprehensive clinical genomic profiling

Katherine Tarlock; Shan Zhong; Yuting He; Rhonda E. Ries; Eric A. Severson; Mark Bailey; Samantha Morley; Sohail Balasubramanian; Rachel L. Erlich; Doron Lipson; Geoff Otto; Jo-Anne Vergillo; E. Anders Kolb; Jeffrey S. Ross; Tariq I. Mughal; Philip J. Stephens; Vincent A. Miller; Soheil Meshinchi; Jie He

Large scale comprehensive genomic profiling (CGP) has led to an improved understanding of oncogenic mutations in acute myeloid leukemia (AML), as well as identification of alterations that can serve as targets for potential therapeutic intervention. We sought to gain insight into age-associated variants in AML through comparison of extensive DNA and RNA-based GP results from pediatric and adult AML. Sequencing of 932 AML specimens (179 pediatric (age 0–18), 753 adult (age ≥ 19)) from diagnostic, relapsed, and refractory times points was performed. Comprehensive DNA (405 genes) and RNA (265) sequencing to identify a variety of structural and short variants was performed. We found that structural variants were highly prevalent in the pediatric cohort compared to the adult cohort (57% vs. 30%; p < 0.001), with certain structural variants detected only in the pediatric cohort. Fusions were the most common structural variant and were highly prevalent in AML in very young children occurring in 68% of children < 2 years of age. We observed an inverse trend in the prevalence of fusions compared to the average number of mutations per patient. In contrast to pediatric AML, adult AML was marked by short variants and multiple mutations per patient. Mutations that were common in adult AML were much less common in the adolescent and young adult cohort and were rare or absent in the pediatric cohort. Clinical CGP demonstrates the biologic differences in pediatric vs. adult AML that have significant therapeutic impacts on prognosis, therapeutic allocation, disease monitoring, and the use of more targeted therapies.


Cancer Research | 2014

Abstract 1658: Enhanced shedding of extracellular vesicles from amoeboid prostate cancer cells: Potential effects on the tumor microenvironment

Jayoung Kim; Samantha Morley; Minh T. N. Le; Denis Bedoret; Dale T. Umetsu; Dolores Di Vizio; Michael R. Freeman

The gene encoding the cytoskeletal regulator DIAPH3 is lost at high frequency in metastatic prostate cancer, and silencing of DIAPH3 evokes a transition to an amoeboid phenotype in multiple tumor cell backgrounds. This amoeboid transformation is accompanied by increases in plasma membrane deformations (blebbing), cell migration, invasion, and metastasis. DIAPH3 silencing also promotes the shedding of atypically large (>1μm) extracellular vesicles (EV) containing bioactive cargo. Whether loss of DIAPH3 also stimulates the release of bioactive nano-sized EV (exosomes) is not established. Here we examined the mechanism of release and potential biological functions of EV shed from DIAPH3-silenced and other prostate cancer cells. We observed that stimulation of LNCaP cells with the prostate stroma-derived growth factor heparin-binding EGF-like growth factor (HB-EGF), combined with p38MAPK inhibition caused exosome shedding, a process mediated by ERK1/2 hyperactivation. DIAPH3 silencing in DU145 cells also increased rates of exosome production. EV isolated from DIAPH3-silenced cells activated AKT1 and androgen signaling, increased proliferation of recipient tumor cells, and suppressed proliferation of human macrophages and peripheral blood mononuclear cells (PBMC). DU145-derived EV contained miR-125a, which suppressed AKT1 expression and proliferation in recipient human peripheral blood mononuclear cells and macrophages. Our findings suggest that EV across a wide size range (∼70nm to >1um) are produced by amoeboid prostate cancer cells as a result of DIAPH3 loss and/or growth factor stimulation. These EV may condition the tumor microenvironment through multiple mechanisms, including promoting the growth of cancer cells and suppression of tumor-infiltrating immune cells. Citation Format: Jayoung Kim, Samantha Morley, Minh Le, Denis Bedoret, Dale Umetsu, Dolores Di Vizio, Michael Freeman. Enhanced shedding of extracellular vesicles from amoeboid prostate cancer cells: Potential effects on the tumor microenvironment. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1658. doi:10.1158/1538-7445.AM2014-1658


Cancer Research | 2014

Abstract 4868: Large oncosomes are internalized and functionally modulate transcription factors in recipient cells

Valentina R. Minciacchi; Matteo Morello; Sungyong You; Wei Yang; Mariana Sobreiro; Cristiana Spinelli; Mandana Zandian; Mirja Rotinen; Samantha Morley; Rosalyn M. Adam; Michael R. Freeman; Dolores Di Vizio

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CA Background and Objective: We recently reported that rapidly migratory, “amoeboid” prostate cancer (PCa) cells shed large (1-10µm diameter), bioactive extracellular vesicles (EV), termed large oncosomes (LO), whose abundance correlates with tumor aggressiveness (Di Vizio et al., Cancer Res. 2009; Di Vizio et al. Am J Pathol. 2012). Increasing evidence supports an important role for EVs in mechanisms of communication between cancer cells and the surrounding microenvironment. EVs can activate signal transduction as well as transfer biomolecules to recipient cells, processes that may promote oncogenesis and/or enhance tumor progression. The aim of this study was to investigate the molecular mechanisms of LO internalization into recipient cells and LO-mediated intercellular communication. Results: LO stimulated the migration of tumor and endothelial cells and contained active MMP2 and MMP9, key proteases involved in tumor cell invasion. Quantitative LC-MS/MS SILAC analysis of LO and exosomes demonstrated enrichment of specific proteins in LO in comparison with exosomes. Fluorescently labeled-LO were internalized into recipient cells and maintained their stability as discrete microvesicles that were localized in the perinuclear space at early time points and into the nucleus at later times. Because of this transfer of LO to the nuclear compartment, we investigated whether exposure to LO altered the activity of transcription factors in recipient cells, hypothesizing a possible novel mechanism of LO-mediated intracellular communication. Interestingly, the degree of internalization, as quantitatively assessed by flow cytometry, varied among different recipient cells, including benign and cancer prostate epithelial cells, immortalized myofibroblasts, cancer associated fibroblasts and endothelial cells. These results suggest cell-specific affinities for target cells and, perhaps, finely regulated mechanisms underlying LO-internalization. Moreover selective inhibition of pathways important in EV internalization indicated that the LO enter cells through an active endocytic process, rather than passively fusing with the plasma membrane of recipient cells. Internalization of LO resulted in altered expression of transcription factors. Conclusions: Our results show for the first time that internalization of intact LO might be necessary for their biological activity in recipient cells. These findings also suggest that the functional role of LO in the tumor microenvironment might be mediated by modulation of transcription factors. Citation Format: Valentina R. Minciacchi, Matteo Morello, Sungyong You, Wei Yang, Mariana Sobreiro, Cristiana Spinelli, Mandana Zandian, Mirja Rotinen, Samantha Morley, Rosalyn Adam, Michael R. Freeman, Dolores Di Vizio. Large oncosomes are internalized and functionally modulate transcription factors in recipient cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4868. doi:10.1158/1538-7445.AM2014-4868

Collaboration


Dive into the Samantha Morley's collaboration.

Top Co-Authors

Avatar

Michael R. Freeman

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Dolores Di Vizio

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Sungyong You

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Rosalyn M. Adam

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Jayoung Kim

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Matteo Morello

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Martin H. Hager

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Wei Yang

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Beatrice Knudsen

Cedars-Sinai Medical Center

View shared research outputs
Top Co-Authors

Avatar

Mirja Rotinen

Cedars-Sinai Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge